close

Immune and Hematology System

Immune and Hematology SystemNatural RemediesQuestions and AnswersSupplementsVitamins and Minerals

Vitamins for immune system

Vitamins for immune system

Immune system

The immune system is made up of a complex network of cells, chemicals, tissues, organs, and the substances they make that helps your body fight infections and other diseases. The immune system includes white blood cells and organs and tissues of the lymph system, such as the thymus, spleen, tonsils, lymph nodes, lymph vessels, and bone marrow (see Figures 1 and 2 below). The immune system must recognize foreign invaders and abnormal cells (Table 1) and distinguish them from the body’s healthy cells. An underactive or overactive immune system can cause health issues. For example, autoimmune diseases, such as multiple sclerosis and rheumatoid arthritis, happen when the body mounts an immune response against its own tissues instead of a foreign invader. In addition, allergies occur when an individual’s immune system reacts to substances in the environment that are tolerated by most people. Underactivity of the immune system or immunodeficiency, can increase your risk of infection. You may be born with an immunodeficiency (known as primary immunodeficiency or inborn errors of immunity), or acquire it from a medical treatment or another disease (known as secondary immunodeficiency). Primary immunodeficiency are a group of more than 450 rare, chronic conditions in which part of the body’s immune system is missing or does not function correctly. Primary immunodeficiencies are caused by hereditary genetic defects and can affect anyone, regardless of age, gender, or ethnicity 1. Because the most important function of the immune system is to protect against infection, children and adults with primary immunodeficiency commonly experience increased susceptibility to infection. The infections may be in the skin, sinuses, throat, ears, lungs, brain or spinal cord, or in the urinary or intestinal tracts. Increased susceptibility to infection may show up as repeated infections, infections that are unusually hard to cure or unusually severe infections (requires hospitalization or intravenous antibiotics).

The immune system is constantly working to protect your body from infections, injury, cancers and diseases. The immune system recognizes invaders such as bacteria, viruses and fungi as well as abnormal cells. It mounts an immune response to help the body fight the invasion. When harmful microbes (tiny particles) enter and invade your body, the body produces white blood cells to fight the infection. The white blood cells identify the microbe, produce antibodies (immunoglobulins or or gamma-globulins) to fight it, and help other immune responses to occur. They also ‘remember’ the attack. There are five major classes of antibodies (IgG, IgA, IgM, IgD, and IgE). IgG has four different subclasses (IgG1, IgG2, IgG3, IgG4). IgA has two subclasses (IgA1 and IgA2). Antibodies of the IgA class are produced near mucus membranes and find their way into secretions such as tears, intestines, bile, saliva and mucus, where they protect against infection in the respiratory tract and intestines. Antibodies of the IgM class are the first antibodies formed in response to infection. They are important in protection during the early days of an infection. Antibodies of the IgE class are responsible for allergic reactions. IgD is expressed on mature B cells along with IgM and may play some role in helping B cells differentiate into plasma cells. Recently, studies have suggested that IgD may be important in the gut homeostasis by binding to mast cells and basophils to react against pathogenic bacteria in the gut.

Antibodies protect the body against infection in a number of different ways. For example, some microorganisms, such as viruses, must attach to body cells before they can cause an infection, but antibodies bound to the surface of a virus can interfere with the virus’ ability to attach to the host cell. In addition, antibodies attached to the surface of some microorganisms can cause the activation of a group of proteins called the complement system that can directly kill some bacteria. Antibody-coated bacteria are also much easier for neutrophils to ingest and kill than bacteria that are not coated with antibodies. All of these actions of antibodies prevent microorganisms from successfully invading body tissues and causing serious infections.

The immune response is split into two functional divisions: innate and acquired immunity.

  1. Innate immunity is the first line of defense against foreign invaders. Innate immunity involves immediate, nonspecific responses to pathogens.
  2. Acquired immunity also called adaptive immunity, is the second line of defense against foreign invaders. Acquired immunity involves a complex, targeted response to a specific pathogen. Exposure to a pathogen stimulates the production of certain immune cells that mark the pathogen for destruction. Upon first exposure, it takes several days or weeks to develop the acquired immune response, but the involved immune cells “remember” the encounter and respond quickly upon subsequent exposure to the same pathogen.

The components of the innate and acquired immune systems communicate and work together to protect your body from infection and disease (Table 2).

Major organs of the Immune System (Figure 1):

  • Thymus: The thymus is an organ located in the upper chest where T cells mature. First, lymphocytes (a type of white blood cell) that are destined to become T cells leave the bone marrow and find their way to the thymus where they are then “educated” to become mature T cells.
  • Liver: The liver is the major organ responsible for producing proteins of the complement system. In addition, it contains large numbers of phagocytic cells (a specific type of white blood cell) that ingest bacteria in the blood as it passes through the liver.
  • Bone Marrow: The bone marrow is the location where all cells of the immune system begin their development from stem cells.
  • Tonsils: Tonsils are collections of lymphocytes in the throat.
  • Lymph Nodes: Lymph nodes are collections of B cells and T cells throughout the body. Cells congregate in lymph nodes to communicate with each other. Lymph nodes can become swollen when they are fighting an infection.
  • Spleen: The spleen is a collection of B cells, T cells, and monocytes. It serves to filter the blood and provide a site for invaders/germs and cells of the immune system to interact.
  • Blood: Blood is contained within the circulatory system that carries cells and proteins of the immune system from one part of the body to another.

The cells that are involved in the immune system (Figure 2):

  • Granulocytes include basophils, eosinophils, and neutrophils. Basophils and eosinophils are important for host defense against parasites. They also are involved in allergic reactions.
  • Neutrophils (also known as polymorphonuclear cells, PMNs or granulocytes), the most numerous innate immune cell (the most numerous of all the types of white blood cells), patrol for problems by circulating in the bloodstream. Neutrophils or polymorphonuclear leukocytes are found in the bloodstream and can migrate into sites of infection within a matter of minutes. These cells, like the other cells in the immune system, develop from hematopoietic stem cells in the bone marrow. Neutrophils can phagocytose, or ingest, bacteria, degrading them inside special compartments called vesicles. Neutrophils increase in number in the bloodstream during infection and are in large part responsible for the elevated white blood cell count seen with some infections. They are the cells that leave the bloodstream and accumulate in the tissues during the first few hours of an infection and are responsible for the formation of pus. Their major role is to ingest bacteria or fungi and kill them. Their killing strategy relies on ingesting the infecting organisms in specialized pockets within the cell. Neutrophils contain toxic chemicals that fuse with the bacteria-containing pockets to kill the bacteria. Neutrophils have little role in the defense against viruses.
  • Monocytes, which develop into macrophages, also patrol and respond to problems. Monocytes make up 5 to 10% of the white blood cells. They also line the walls of blood vessels in organs like the liver and spleen where they capture microorganisms in the blood as they pass by. When monocytes leave the bloodstream and enter the tissues, they change shape and size and become macrophages. Macrophages, “big eater” in Greek, are named for their ability to ingest and degrade bacteria. Macrophages are essential for killing fungi and the class of bacteria to which tuberculosis belongs (mycobacteria). Like neutrophils, macrophages ingest microbes and deliver toxic chemicals directly to the foreign invader to kill it. Macrophages live longer than neutrophils and are especially important for slow growing or chronic infections. Macrophages can be influenced by T cells and often collaborate with T cells in killing microorganisms. Macrophages also have important non-immune functions, such as recycling dead cells, like red blood cells, and clearing away cellular debris. These “housekeeping” functions occur without activation of an immune response. Upon activation, monocytes and macrophages coordinate an immune response by notifying other immune cells of the problem.
  • B cells (B lymphocytes) and often named on lab reports as CD19 or CD20 cells: These lymphocytes arise in the bone marrow from stem cells. When B cells encounter foreign germs (antigens), they respond by maturing into another cell type called plasma cells. Plasma cells are the mature B cells (mature B lymphocytes) that actually produce the antibodies (also known as immunoglobulins or gamma-globulins) and are located in the spleen and lymph nodes throughout the body. B cells can also mature into memory cells, which allows a rapid response if the same infection is encountered again. The long life of plasma cells enables your body to retain immunity to viruses and bacteria that infected you many years ago. For example, once you have been fully immunized with live vaccine strains of measles virus, you will almost never catch it because your body retain the plasma cells and antibodies for many years and these antibodies prevent infection.
  • T cells sometimes called T lymphocytes and often named in lab reports as CD3 cells, are another type of immune cell. Some T cells directly attack cells infected with viruses, and others act as regulators of the immune system. Each T cell reacts with one specific antigen, just as each antibody molecule reacts with one specific antigen. In fact, T cells have molecules on their surfaces that are similar to antibodies. The variety of different T cells is also so extensive that the body has T cells that can react against virtually any antigen. T cells have different abilities to recognize antigen and are varied in their function. There are killer or cytotoxic T cells (often denoted in lab reports as CD8 T cells), helper T cells (often denoted in lab reports as CD4 T cells), and regulatory T cells. Each has a different role to play in the immune system.
  • Cytotoxic T cells (CD8+ T cells or Killer T cells): These lymphocytes mature in the thymus and are responsible for killing cells infected with viruses. Killer T cells protect the body from certain bacteria and viruses that have the ability to survive and even reproduce within the body’s own cells. The killer T cell must migrate to the site of infection and directly bind to its target to ensure its destruction. In addition to fighting germs, killer T cells also recognize and respond to foreign tissues in the body, such as a transplanted kidney. When T cells are fighting infections, they grow and divide, making more T cells.
  • Helper T cells (CD4+ T-cell): Helper T cells assist B cells to produce antibodies and assist killer T cells in their attack on foreign substances.
  • Regulatory T cells. Regulatory T cells suppress or turn off the T cells when an infection is controlled and they are no longer needed. Regulatory T cells act as the thermostat of the lymphocyte system to keep it turned on just enough—not too much and not too little. Without regulatory T cells, the immune system would keep working even after an infection has been treated. Without regulatory T cells, there is the potential for the body to overreact to the infection.
  • Plasma cells: These cells develop from B cells (B lymphocytes) and are the cells that make immunoglobulin (antibodies).
  • Mast cells also are important for defense against parasites. Mast cells are found in tissues and can mediate allergic reactions by releasing inflammatory chemicals like histamine.
  • Dendritic cells (DC) also known as antigen-presenting cells (APCs), instruct T cells on what to attack. Dendritic cells (DC) also can develop from monocytes. Antigens are molecules from pathogens, host cells, and allergens that may be recognized by adaptive immune cells. APCs like DCs are responsible for processing large molecules into “readable” fragments (antigens) recognized by adaptive B or T cells. However, antigens alone cannot activate T cells. They must be presented with the appropriate major histocompatiblity complex (MHC) expressed on the APC. MHC provides a checkpoint and helps immune cells distinguish between host and foreign cells.
  • Natural killer (NK) cells have features of both innate and adaptive immunity. They are important for recognizing and killing virus-infected cells or tumor cells. Natural killer (NK) cells are so named because they easily kill cells infected with viruses. They are said to be natural killer cells as they are always ready to fight and do not require the same thymus education that T cells require. NK cells are derived from the bone marrow and are present in relatively low numbers in the bloodstream and in tissues. They are important in defending against viruses and possibly preventing cancer as well. They contain intracellular compartments chemicals called cytotoxic granules, which are filled with proteins that can form holes in the target cell and also cause apoptosis, the process for programmed cell death. NK cells kill virus-infected cells by injecting them with a killer potion of chemicals called cytotoxic granules. It is important to distinguish between apoptosis and other forms of cell death like necrosis. Apoptosis, unlike necrosis, does not release danger signals that can lead to greater immune activation and inflammation. Through apoptosis, immune cells can discreetly remove infected cells and limit bystander damage. NK cells are particularly important in the defense against herpes viruses. This family of viruses includes the traditional cold sore form of herpes (herpes simplex) as well as Epstein-Barr virus (the cause of infectious mononucleosis or mono) and the varicella virus (the cause of chickenpox and shingles). Recently, researchers have shown in mouse models that NK cells, like adaptive cells, can be retained as memory cells and respond to subsequent infections by the same pathogen.

The immune system relies on an adequate supply of nutrients for its baseline functions as well as for ramping up its activity when necessary 2, 3, 4, 5, 6, 7. It is well established that malnutrition (protein-energy malnutrition and obesity) and deficiencies in one or more micronutrients (vitamins and nutritionally essential minerals) diminish immune function. In most instances, correcting the nutrient deficiency restores the affected immune functions. At a minimum, getting the recommended dietary allowance (RDA) for vitamin C and vitamin D is necessary for the immune system to function properly; there is some evidence that intakes above the current RDA (recommended dietary allowance) for these vitamins may be of further benefit 8. Because supplementation with iron can have unwanted side effects in those with preexisting infections, especially malaria, routine iron supplementation should be accompanied by malaria detection and treatment strategies 8. The long-chain Omega-3 Polyunsaturated Fatty Acids (PUFA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have potent anti-inflammatory effects, especially in individuals with chronic or acute inflammation 8. Increasing eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) consumption increases the EPA and DHA content of immune cell membranes, mainly by displacing the long-chain Omega-6 Polyunsaturated Fatty Acids (PUFA) arachidonic acid and becoming the substrate for the enzymes that synthesize eicosanoids. EPA and DHA also give rise to anti-inflammatory compounds that “turn off” the inflammatory response. In chronic inflammatory states (e.g., rheumatoid arthritis, atherosclerosis), eicosapentaenoic acid (EPA) plus docosahexaenoic acid (DHA) supplements have been shown to reduce symptoms of rheumatoid arthritis and the risk of cardiac events. However, the dose of EPA plus DHA that is optimal for immune function in healthy individuals is not yet established. Ingestion of at least 500 mg/day of EPA plus DHA is recommended by the International Society for the Study of Fatty Acids and Lipids 9 and the American Heart Association recommends eating one to two servings of seafood per week to reduce your risk of some heart problems, especially if you consume the seafood in place of less healthy foods 10. For people with heart disease, the American Heart Association recommends consuming about 1 g per day EPA plus DHA, preferably from oily fish, but supplements are an option under the guidance of a health care provider 11. The AHA does not recommend omega-3 supplements for people who do not have a high risk of cardiovascular disease 12. The Linus Pauling Institute recommends that generally healthy adults eat fish twice weekly, which provides approximately 500 mg/day of EPA plus DHA; for those who do not regularly consume fish, consider taking a two-gram fish oil supplement several times a week in consultation with a physician 8. Higher daily intakes may be recommended for the treatment of specific disorders.

Table 1.  The Immune System responds to Foreign Invaders and Abnormal Cells

Foreign InvadersAbnormal Cells
VirusesCancer cells
Bacteria
Parasites
Mold
Allergens
[Source 8 ]

Table 2. Functional Divisions and Components of the Immune Response

Innate Immunity (general response to a pathogen)Acquired Immunity (specific response to a pathogen)
BarriersCellsHumoral FactorsCellsHumoral Factors
Physical: skin, mucous membranesPhagocytes: engulf and destroyEicosanoids: Eicosanoids are compounds made from 20-carbon long-chain polyunsaturated fatty acids (PUFA); the term ‘eicosanoid’ includes many compounds that can either cause, prevent or regulate inflammationT lymphocytes: detect specific pathogens, secrete cytokines, and coordinate an immune responseAntibodies: specialized proteins that mark a pathogen for destruction
Chemical: acidic environment of the stomachMast cells: cause inflammation & symptoms of allergyCytokines: Cytokines are proteins made by white blood cells. Cytokines regulate inflammation. They play important roles in your body’s normal immune responses and in the immune system’s ability to respond to cancer.
B lymphocytes: produce antibodies against a specific pathogen
Biological: gut microbiotaNatural killer (NK) cells: release toxic chemicalsComplement proteins: attach to and destroy bacteria; some cause inflammationComplement proteins: attach to and destroy pathogens marked by antibodies
[Source 8 ]

Figure 1. Immune system

the immune system

Figure 2. Cells of the immune system

Footnote: The cells of the immune system originate in the bone marrow from pluripotent hematopoietic stem cells. Pluripotent hematopoietic stem cells give rise to a common lymphoid progenitor, which gives rise to all of the major lymphoid cell types (T‐cells, B‐cells, and Natural killer [NK] cells) or a common myeloid progenitor, which gives rise to all of the major myeloid cell types (neutrophils, eosinophils, basophils, dendritic cells (DCs), mast cells, and monocytes/macrophages) as well as the erythrocytes and megakaryocytes (which generate platelets).

Figure 3. Factors that influence the immune response

Factors that influence the immune response

[Source 5 ]

Figure 4. Vitamins and minerals for immune system

Vitamins and minerals for immune system

Footnotes: Vitamins and minerals have key roles at every stage of the immune response 13. This schematic summarizes important components and processes that are involved in different aspects of the innate and adaptive immune responses. The circles highlight those micronutrients that are known to affect these responses. The significant overlap between micronutrients and processes indicates the importance of multiple micronutrients in supporting proper function of the immune system.

Abbreviations: APCs = antigen-presenting cells; C3 = complement component 3; CRP = C-reactive protein; Cu = copper; Fe = iron; IFNs = interferons; Igs = immunoglobulins; ILs = interleukins; GI = gastrointestinal; GM-CSF = granulocyte-macrophage colony stimulating factor; MAC = membrane attack complex; MCP-1 = monocyte chemoattractant protein-1; Mg = magnesium; MHCs = major histocompatibility complexes; NK = natural killer; NO = nitric oxide; ROS = reactive oxygen species; Se = selenium; TLRs = toll-like receptors; TNF = tumor-necrosis factors; Zn = zinc

[Source 7 ]

How does the immune system work?

The immune system involves many parts of your body. Each part plays a role in recognizing foreign microbes, communicating with other parts of your body, and working to fight the infection. Parts of the immune system are:

  • Skin – The skin is usually the first line of defense against microbes. Skin cells produce and secrete important antimicrobial proteins, and immune cells can be found in specific layers of skin.
  • Bone marrow – helps produce immune cells. The bone marrow contains stems cells that can develop into a variety of cell types. The common myeloid progenitor stem cell in the bone marrow is the precursor to innate immune cells—neutrophils, eosinophils, basophils, mast cells, monocytes, dendritic cells, and macrophages—that are important first-line responders to infection. The common lymphoid progenitor stem cell leads to adaptive immune cells—B cells and T cells—that are responsible for mounting responses to specific microbes based on previous encounters (immunological memory). Natural killer (NK) cells also are derived from the common lymphoid progenitor and share features of both innate and adaptive immune cells, as they provide immediate defenses like innate cells but also may be retained as memory cells like adaptive cells. B, T, and NK cells also are called lymphocytes.
  • Bloodstream: Immune cells constantly circulate throughout the bloodstream, patrolling for problems. When blood tests are used to monitor white blood cells, another term for immune cells, a snapshot of the immune system is taken. If a cell type is either scarce or overabundant in the bloodstream, this may reflect a problem.
  • The thymus, a small gland in your upper chest where some immune T cells mature.
  • Lymphatic system is a network of of tiny vessels which allows immune cells to travel between tissues and the bloodstream. The lymphatic system contains lymphocytes (white blood cells; mostly T cells and B cells), which try to recognize any bacteria, viruses or other foreign substances in the body and fight them. They are carried in a milky fluid called lymph. Immune cells are carried through the lymphatic system and converge in lymph nodes, which are found throughout the body.
  • Lymph nodes, small lumps in the groin, armpit, around the neck and elsewhere that help the lymphatic system to communicate. Lymph nodes are a communication hub where immune cells sample information brought in from the body. They can become swollen when the body mounts an immune response. For instance, if adaptive immune cells in the lymph node recognize pieces of a microbe brought in from a distant area, they will activate, replicate, and leave the lymph node to circulate and address the pathogen. Thus, doctors may check patients for swollen lymph nodes, which may indicate an active immune response.
  • The spleen is an organ under the ribs behind the stomach on the left that processes information from the blood. While it is not directly connected to the lymphatic system, it is important for processing information from the bloodstream. Immune cells are enriched in specific areas of the spleen, and upon recognizing blood-borne pathogens, they will activate and respond accordingly.
  • Mucous membranes, like the lining of the inside of your mouth are prime entry points for pathogens, and specialized immune hubs are strategically located in mucosal tissues like the respiratory tract and gut. For instance, Peyer’s patches are important areas in the small intestine where immune cells can access samples from the gastrointestinal tract.​

Immune organs

The organ systems involved in the immune response are primarily lymphoid organs which include, spleen, thymus, bone marrow, lymph nodes, tonsils, and liver. The lymphoid organ system classifies according to the following 14:

  1. Primary lymphoid organs (thymus and bone marrow), where T and B cells first express antigen receptors and become mature functionally.
  2. Secondary lymphoid organs like the spleen, tonsils, lymph nodes, the cutaneous and mucosal immune system; this is where B and T lymphocytes recognize foreign antigens and develop appropriate immune responses.

All immune cells originate in the bone marrow, deriving from hematopoietic stem cells, but an important set of immune cells (T lymphocytes) undergo maturation in an organ known as the thymus. The thymus and bone marrow are known as primary lymphoid tissues. T lymphocytes mature in the thymus, where these cells reach a stage of functional competence while B lymphocytes mature in the bone marrow the site of generation of all circulating blood cells. Excessive release of cytokines stimulated by these organisms can cause tissue damage, such as endotoxin shock syndrome.

Secondary lymphoid tissues, namely the lymph nodes, spleen and mucosa-associated lymphoid tissues (MALT) are important sites for generating adaptive immune responses and contain the lymphocytes (key adaptive cells). The lymphatic system is a system of vessels draining fluid (derived from blood plasma) from body tissues. Lymph nodes, that house lymphocytes, are positioned along draining lymph vessels, and monitor the lymph for signs of infection. MALT tissues are important in mucosal immune responses, and reflect the particular importance of the gut and airways in immune defence. The spleen essentially serves as a ‘lymph node’ for the blood.

Immune cells communication

Immune cells communicate in a number of ways, either by cell-to-cell contact or through secreted signaling molecules. Receptors and ligands are fundamental for cellular communication.

  • Receptors are protein structures that may be expressed on the surface of a cell or in intracellular compartments. The molecules that activate receptors are called ligands, which may be free-floating or membrane-bound.
  • Ligand-receptor interaction leads to a series of events inside the cell involving networks of intracellular molecules that relay the message. By altering the expression and density of various receptors and ligands, immune cells can dispatch specific instructions tailored to the situation at hand.

Cytokines are small proteins with diverse functions. In immunity, there are several categories of cytokines important for immune cell growth, activation, and function.

  • Colony-stimulating factors are essential for cell development and differentiation.
  • Interferons (IFNs) are necessary for immune-cell activation. Type I interferons mediate antiviral immune responses, and type II interferon is important for antibacterial responses.
  • Interleukins (ILs), which come in over 30 varieties, provide context-specific instructions, with activating or inhibitory responses.
  • Chemokines are made in specific locations of the body or at a site of infection to attract immune cells. Different chemokines will recruit different immune cells to the site needed.
  • Tumor necrosis factor (TNF) family of cytokines stimulates immune-cell proliferation and activation. They are critical for activating inflammatory responses, and as such, TNF blockers are used to treat a variety of disorders, including some autoimmune diseases.

Toll-like receptors (TLRs) are expressed on innate immune cells, like macrophages and dendritic cells. They are located on the cell surface or in intracellular compartments because microbes may be found in the body or inside infected cells. TLRs recognize general microbial patterns, and they are essential for innate immune-cell activation and inflammatory responses.

B-cell receptors (BCRs) and T-cell receptors (TCRs) are expressed on adaptive immune cells. They are both found on the cell surface, but BCRs also are secreted as antibodies to neutralize pathogens. The genes for BCRs and TCRs are randomly rearranged at specific cell-maturation stages, resulting in unique receptors that may potentially recognize anything. Random generation of receptors allows the immune system to respond to unforeseen problems. They also explain why memory B or T cells are highly specific and, upon re-encountering their specific pathogen, can immediately induce a neutralizing immune response.

Major histocompatibility complex (MHC) or human leukocyte antigen (HLA), proteins serve two general roles.

Major histocompatibility complex (MHC) proteins function as carriers to present antigens on cell surfaces. MHC class I proteins are essential for presenting viral antigens and are expressed by nearly all cell types, except red blood cells. Any cell infected by a virus has the ability to signal the problem through MHC class I proteins. In response, CD8+ T cells (also called CTLs) will recognize and kill infected cells. MHC class II proteins are generally only expressed by antigen-presenting cells like dendritic cells and macrophages. MHC class II proteins are important for presenting antigens to CD4+ T cells. MHC class II antigens are varied and include both pathogen- and host-derived molecules.

MHC proteins also signal whether a cell is a host cell or a foreign cell. They are very diverse, and every person has a unique set of MHC proteins inherited from his or her parents. As such, there are similarities in MHC proteins between family members. Immune cells use MHC to determine whether or not a cell is friendly. In organ transplantation, the MHC or HLA proteins of donors and recipients are matched to lower the risk of transplant rejection, which occurs when the recipient’s immune system attacks the donor tissue or organ. In stem cell or bone marrow transplantation, improper MHC or HLA matching can result in graft-versus-host disease, which occurs when the donor cells attack the recipient’s body.

Complement refers to a unique process that clears away pathogens or dying cells and also activates immune cells. Complement consists of a series of proteins found in the blood that form a membrane-attack complex. Complement proteins are only activated by enzymes when a problem, like an infection, occurs. Activated complement proteins stick to a pathogen, recruiting and activating additional complement proteins, which assemble in a specific order to form a round pore or hole. Complement literally punches small holes into the pathogen, creating leaks that lead to cell death. Complement proteins also serve as signaling molecules that alert immune cells and recruit them to the problem area.

Best vitamins for immune system

Through experimental research and studies of people with immune deficiencies, a number of vitamins (A, B6, B12, folate, C, D and E) and trace elements (zinc, copper, selenium, iron) have been demonstrated to have key roles in supporting the human immune system and reducing risk of infections 15. Other essential nutrients including other vitamins and trace elements, amino acids and fatty acids are also important in this regard. All of the nutrients named above have roles in supporting antibacterial and antiviral defences but zinc and selenium seem to be particularly important for the latter.

Table 3. Vitamins and minerals that affect the immune system

NameWhat It DoesWhere to Get ItAbout Supplements
Vitamin A (Retinol) and beta-caroteneKeeps skin, lungs, and stomach healthy.

  • Role in barrier function:
    • Promotes differentiation of epithelial tissue; promotes gut homing of B- and T cells; promotes intestinal immunoglobulin A cells; promotes epithelial integrity
  • Role in cellular aspects of innate immunity:
    • Regulates number and function of NK cells; supports phagocytic and oxidative burst activity of macrophages
  • Role in B-cell mediated immunity:
    • Supports function of B cells; required for immunoglobulin A production
  • Role in T-cell mediated immunity:
    • Regulates development and differentiation of Th1 and Th2 cells; promotes conversion of naive T cells to regulatory T cells; regulates IL-2, IFN-γ and TNF production
liver, whole eggs, milk, dark green, yellow, orange, and red vegetables and fruit (like spinach, pumpkin, green peppers, squash, carrots, papaya, and mangoes). Also found in orange and yellow sweet potatoesIt’s best to get vitamin A from food. Vitamin A supplements are toxic in high doses. Supplements of beta-carotene (the form of vitamin A in fruits and vegetables) have been shown to increase cancer risk in smokers.
Vitamin B-group
Thiamin (Vitamin B1), Riboflavin (Vitamin B2), Vitamin B6, Folate (Vitamin B9), Vitamin B12
Keeps the immune and nervous system healthy.

Vitamin B6

  • Role in barrier function:
    • Promotes gut homing of T cells
  • Role in cellular aspects of innate immunity:
    • Supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Supports antibody production
  • Role in T-cell mediated immunity:
    • Promotes T-cell differentiation, proliferation and function, especially Th1 cells; regulates (promotes) IL-2 production

Folate (Vitamin B9)

  • Role in barrier function:
    • Survival factor for regulatory T cells in the small intestine
  • Role in cellular aspects of innate immunity:
    • Supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Supports antibody production
  • Role in T-cell mediated immunity:
    • Promotes proliferation of T cells and the Th1-cell response

Vitamin B12

  • Role in barrier function:
    • Important co-factor for gut microbiota
  • Role in cellular aspects of innate immunity:
    • Supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Required for antibody production
  • Role in T-cell mediated immunity:
    • Promotes T-cell differentiation, proliferation and function, especially cytotoxic T cells; controls ratio of T-helper to cytotoxic T cells
white beans, potatoes, meat, fish, chicken, watermelon, grains, nuts, avocados, broccoli, and green leafy vegetables
Vitamin CHelps protect the body from infection and aids in recovery.

  • Role in barrier function:
    • Promotes collagen synthesis; promotes keratinocyte differentiation; protects against oxidative damage; promotes wound healing; promotes complement
  • Role in cellular aspects of innate immunity:
    • Supports function of neutrophils, monocytes and macrophages including phagocytosis; supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Promotes antibody production
  • Role in T-cell mediated immunity:
    • Promotes production, differentiation and proliferation of T cells especially cytotoxic T cells; regulates IFN-γ production
citrus fruits (like oranges, grapefruit, and lemons), tomatoes, and potatoes
Vitamin DImportant for developing and maintaining healthy bones and teeth.

  • Role in barrier function:
    • Promotes production of antimicrobial proteins (cathelicidin, β-defensin); promotes gut tight junctions (via E-cadherin, connexion 43); promotes homing of T cells to the skin
  • Role in cellular aspects of innate immunity:
    • Promotes differentiation of monocytes to macrophages; promotes macrophage phagocytosis and oxidative burst
  • Role in B-cell mediated immunity:
    • Can decrease antibody production
  • Role in T-cell mediated immunity:
    • Promotes antigen processing but can inhibit antigen presentation; can inhibit T-cell proliferation, Th1-cell function and cytotoxic T-cell function; Promotes the development of regulatory T cells; inhibits differentiation and maturation of dendritic cells; regulates IFN-γ production
fortified milk, fatty fish, sunlight
Vitamin EProtects cells and helps fight off infection.

  • Role in barrier function:
    • Protects against oxidative damage
  • Role in cellular aspects of innate immunity:
    • Supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Supports antibody production
  • Role in T-cell mediated immunity:
    • Promotes interaction between dendritic cells and T cells; promotes T-cell proliferation and function, especially Th1 cells; regulates (promotes) IL-2 production
green leafy vegetables, vegetable oils, avocados, almondsLimit to 400 IU per day.
IronNot having enough iron can cause anemia.

  • Role in barrier function:
    • Essential for growth and differentiation of epithelial tissue
  • Role in cellular aspects of innate immunity:
    • Promotes bacterial killing by neutrophils; regulates balance of M1 and M2 macrophages; supports NK-cell activity
  • Role in T-cell mediated immunity:
    • Regulates differentiation and proliferation of T cells; regulates IFN-γ production
green leafy vegetables, whole grain breads and pastas, dried fruit, beans, red meat, chicken, liver, fish, and eggsLimit to 45 mg per day unless otherwise instructed by your doctor. Iron may be a problem for people with HIV because it can increase the activity of some bacteria. Supplements that do not contain iron may be better.
CopperCopper is a cofactor for several enzymes (known as “cuproenzymes”) involved in energy production, iron metabolism, neuropeptide activation, connective tissue synthesis, and neurotransmitter synthesis. Copper is also involved in many physiologic processes, such as angiogenesis; neurohormone homeostasis; and regulation of gene expression, brain development, pigmentation, and immune system functioning. In addition, defense against oxidative damage depends mainly on the copper-containing superoxide dismutases.

  • Role in cellular aspects of innate immunity:
    • Promotes neutrophil, monocyte and macrophage phagocytosis; supports NK-cell activity
  • Role in T-cell mediated immunity:
    • Regulates differentiation and proliferation of T cells; regulates (promotes) IL-2 production
beef liver, shellfish, seeds and nuts, organ meats, wheat-bran cereals, whole-grain products, and chocolate

Tap water and other beverages can also be sources of copper, although the amount of copper in these liquids varies by source (ranging from 0.0005 mg/L to 1 mg/L)

Limit to 900 mcg per day.
MagnesiumImportant for the immune system.

  • Cofactor of enzymes of nucleic acid metabolism and stabilizes structure of nucleic acids;
  • Increased numbers of monocytes 16;
  • Increased NK-cell activity 16;
  • Decreased oxidative stress after strenuous exercise 16;
  • After exercises, increases granulocyte count and post-exercise lymphopenia 17;
  • Decreased levels of cytokines such as IL-6 18
  • Decreased inflammation 18
  • Increased T-cell ratios 16
  • Involved in DNA replication and repair. Reduces oxidative damage to the DNA of peripheral blood lymphocytes in athletes and sedentary young men 19;
  • Roles in antigen binding to macrophages 18;
  • Regulates leukocyte activation 18;
  • Reduces leukocyte activation 20;
  • Involved in the regulation of apoptosis 19
green leafy vegetables, such as spinach, legumes, nuts, seeds, and whole grains, are good sources of magnesiumLimit 420 mg (male) and 320 mg (female) per day.
SeleniumImportant for the immune system.

  • Role in cellular aspects of innate immunity:
    • Supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Supports antibody production
  • Role in T-cell mediated immunity:
    • Regulates differentiation and proliferation of T cells; regulates (promotes) IFN-γ production
whole grains, meat, fish, poultry, eggs, peanut butter, and nutsLimit to 400 mcg per day.
ZincImportant for the immune system.

  • Role in barrier function:
    • Maintains integrity of the skin and mucosal membranes; promotes complement activity
  • Role in cellular aspects of innate immunity:
    • Supports monocyte and macrophage phagocytosis; supports NK-cell activity
  • Role in B-cell mediated immunity:
    • Supports antibody production particularly immunoglobulin G (Ig G)
  • Role in T-cell mediated immunity:
    • Promotes Th1-cell response; Promotes proliferation of cytotoxic T cells; promotes development of regulatory T cells; regulates (promotes) IL-2 and IFN-γ production; reduces development of Th9 and Th17 cells
meat, fish, poultry, beans, peanuts, and milk and dairy productsLimit to 40 mg per day.

Abbreviations: IFN = Interferon; IL= interleukin; NK = natural killer; Th = T-helper; TNF = tumor necrosis factor

[Source 215 ]

Table 4. Important dietary sources of nutrients that support the immune system

NutrientGood dietary sources
Vitamin A (or equivalents)Milk and cheese, eggs, liver, oily fish, fortified cereals, dark orange or green vegetables (eg, carrots, sweet potatoes, pumpkin, squash, kale, spinach, broccoli), orange fruits (eg, apricots, peaches, papaya, mango, cantaloupe melon), tomato juice
Vitamin B6Fish, poultry, meat, eggs, whole grain cereals, fortified cereals, many vegetables (especially green leafy) and fruits, soya beans, tofu, yeast extract
Vitamin B12Fish, meat, some shellfish, milk and cheese, eggs, fortified breakfast cereals, yeast extract
Folate (Vitamin B9)Broccoli, brussels sprouts, green leafy vegetables (spinach, kale, cabbage), peas, chick peas, fortified cereals
Vitamin COranges and orange juice, red and green peppers, strawberries, blackcurrants, kiwi, broccoli, brussels sprouts, potatoes
Vitamin DOily fish, liver, eggs, fortified foods (spreads and some breakfast cereals)
Vitamin EMany vegetable oils, nuts and seeds, wheat germ (in cereals)
ZincShellfish, meat, cheese, some grains and seeds, cereals, seeded or wholegrain breads
SeleniumFish, shellfish, meat, eggs, some nuts especially brazil nuts
IronMeat, liver, beans, nuts, dried fruit (eg, apricots), wholegrains (eg, brown rice), fortified cereals, most dark green leafy vegetables (spinach, kale)
CopperShellfish, nuts, liver, some vegetables
MagnesiumGreen leafy vegetables, such as spinach, legumes, nuts, seeds, and whole grains
Essential amino acidsMeat, poultry, fish, eggs, milk and cheese, soya, nuts and seeds, pulses
Essential fatty acidsMany seeds, nuts and vegetable oils
Long chain omega-3 fatty acids (eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA])Oily fish

Table 5. Results of multiple scientific studies (meta-analyses) on micronutrients and respiratory infections

MicronutrientSample sizeMain findingsStated conclusion in abstractReference
Vitamin A47 randomized controlled trials (1 223 856 children)Vitamin A did not affect incidence of, or mortality from, respiratory disease;
Note: vitamin A decreased all cause mortality and mortality from diarrhea and decreased incidence of diarrhea and measles
Vitamin A supplementation is associated with a clinically meaningful reduction in morbidity and mortality in children.Imdad et al 22
Vitamin A15 randomized controlled trials (3021 children)Vitamin A did not affect mortality of children with pneumonia.
Vitamin A decreased pneumonia morbidity, increased the clinical response rate, shortened clearance time of signs and shortened length of hospital stay.
Vitamin A supplementation helps to relieve clinical symptoms and signs (of pneumonia) and shorten the length of hospital stay.Hu et al 23
Vitamin C3 prophylactic trials (2335 participants) two therapeutic trials (197 patients)All three trials found vitamin C decreased the incidence of pneumonia. One trial found vitamin C decreased severity and mortality from pneumonia; the other trial found vitamin C shortened duration of pneumonia.Hemila and Louhiala 24
Vitamin C29 prophylactic randomized controlled trials investigating incidence (11 306 participants) 31 prophylactic randomized controlled trials investigating duration (9745 episodes)Vitamin C did not affect incidence of the common cold in the general population (24 randomized controlled trials) but decreased incidence in people under heavy short-term physical stress (5 randomized controlled trials). Vitamin C shortened duration of common cold in all studies (31 randomized controlled trials), in adults (13 randomized controlled trials) and in children (10 randomized controlled trials) and decreased severity of colds.Hemila and Chalker 25
Vitamin D11 randomized controlled trials (5660 participants)Vitamin D decreased the risk of respiratory tract infections.Vitamin D has a positive effect against respiratory tract infections and dosing once daily seems most effective.Bergman et al 26
Vitamin D25 randomized controlled trials (11 321 participants)Vitamin D decreased the risk of acute respiratory tract infection, effects greater in those with low starting statusVitamin D supplementation was safe and it protected against respiratory tract infection.Martineau et al 27
Vitamin D24 studies; 14 included in meta-analysis of risk of acute respiratory tract infections and 5 in the meta-analysis of severitySerum vitamin D was inversely associated with risk and severity of acute respiratory tract infections.There is an inverse non-linear association between 25-hydroxyvitamin D concentration and acute respiratory tract infection.Pham et al 28
Vitamin D8 observational studies (20 966 participants)Participants with vitamin D deficiency had increased risk of community-acquired pneumonia.There is an association between vitamin D deficiency and increased risk of community-acquired pneumonia.Zhou et al 29
Zinc, copper and iron13 studies in Chinese childrenChildren with recurrent respiratory tract infection had lower hair levels of zinc, copper and iron.The deficiency of zinc, copper and iron may be a contributing factor for the susceptibility of recurrent respiratory tract infection in Chinese children.Mao et al 30
Zinc7 randomized controlled trials (575 participants)Zinc shortened duration of common cold.Hemila 31
Zinc17 randomized controlled trials (2121 adults and children)Zinc decreased duration of common cold symptoms overall and in adults but not in children.Oral zinc formulations may shorten the duration of symptoms of the common cold.Science et al 32
Zinc6 randomized controlled trials (5193 children)Zinc decreased incidence of pneumonia.
Zinc decreased prevalence of pneumonia.
Zinc supplementation in children is associated with a reduction in the incidence and prevalence of pneumonia.Lassi et al 33
Zinc6 randomized controlled trials (2216 adults with severe pneumonia)Zinc given as an adjunct therapy decreased mortality.
No effect of zinc on treatment failure or antibiotic treatment.
Zinc given as an adjunct to the treatment of severe pneumonia is effective in reducing mortality.Wang and Song 34

List of foods containing Vitamins and Minerals that Affect the Immune System

Vitamin A

Vitamin A is name of a group of fat-soluble vitamin (retinoids, including retinol, retinal, and retinyl esters) 35, 36, 37, that is naturally present in many foods.

Vitamin A is important for normal vision, gene expression, the immune system, embryonic development, growth, and reproduction. Vitamin A also helps the heart, lungs, kidneys, and other organs work properly 38.

There are two different types of vitamin A 39.

  1. The first type, preformed vitamin A (retinol and its esterified form, retinyl ester), is found in meat (especially liver), poultry, fish, and dairy products.
  2. The second type, provitamin A carotenoids (beta-carotene, alpha-carotene and beta-cryptoxanthin), is found in fruits, vegetables, and other plant-based products (oily fruits and red palm oil). The most common type of provitamin A carotenoids in foods and dietary supplements is beta-carotene (β-carotene). The body converts these plant pigments into vitamin A.

There are a number of reviews of the role of vitamin A and its metabolites (eg, 9-cis-retinoic acid) in immunity and in host susceptibility to infection 40. Vitamin A is important for normal differentiation of epithelial tissue and for immune cell maturation and function. Thus, vitamin A deficiency is associated with impaired barrier function, altered immune responses and increased susceptibility to a range of infections. Vitamin A-deficient mice show breakdown of the gut barrier and impaired mucus secretion (due to loss of mucus-producing goblet cells), both of which would facilitate entry of pathogens. Many aspects of innate immunity, in addition to barrier function, are modulated by vitamin A and its metabolites. Vitamin A controls neutrophil maturation and in vitamin A deficiency blood neutrophil numbers are increased, but they have impaired phagocytic function. Therefore, the ability of neutrophils to ingest and kill bacteria is impaired. Vitamin A also supports phagocytic activity and oxidative burst of macrophages, so promoting bacterial killing. Natural killer cell activity is diminished by vitamin A deficiency, which would impair antiviral defences. The impact of vitamin A on acquired immunity is less clear and may depend on the exact setting and the vitamin A metabolite involved. Vitamin A controls dendritic cell and CD4+ T lymphocyte maturation and its deficiency alters the balance between T helper 1 and T helper 2 lymphocytes. Studies in experimental model systems indicate that the vitamin A metabolite 9-cis retinoic acid enhances T helper 1 responses. Retinoic acid promotes movement (homing) of T lymphocytes to the gut-associated lymphoid tissue. Interestingly, some gut-associated immune cells are able to synthesise retinoic acid. Retinoic acid is required for CD8+ T lymphocyte survival and proliferation and for normal functioning of B lymphocytes including antibody generation. Thus, vitamin A deficiency can impair the response to vaccination, as discussed elsewhere 41. In support of this, vitamin A-deficient Indonesian children provided with vitamin A showed a higher antibody response to tetanus vaccination than seen in vitamin A-deficient children 42. Vitamin A deficiency predisposes to respiratory infections, diarrhoea and severe measles. Systematic reviews and meta-analyses of trials in children with vitamin A report reduced all-cause mortality 22, reduced incidence, morbidity and mortality from measles 22 and from infant diarrhoea 22 and improved symptoms in acute pneumonia 23.

You can get recommended amounts of vitamin A by eating a variety of foods, including the following:

  • Beef liver and other organ meats (but these foods are also high in cholesterol, so limit the amount you eat).
  • Some types of fish, such as salmon.
  • Green leafy vegetables and other green, orange, and yellow vegetables, such as broccoli, carrots, and squash.
  • Fruits, including cantaloupe, apricots, and mangos.
  • Dairy products, which are among the major sources of vitamin A for Americans.
  • Fortified breakfast cereals.

Table 4 suggests many dietary sources of vitamin A. The foods from animal sources contain primarily preformed vitamin A, the plant-based foods have provitamin A, and the foods with a mixture of ingredients from animals and plants contain both preformed vitamin A and provitamin A.

sources of vitamin A

Table 6. Selected Food Sources of Vitamin A

Foodmcg RAE per
serving
IU per
serving
Percent
DV*
Sweet potato, baked in skin, 1 whole1,40328,058561
Beef liver, pan fried, 3 ounces6,58222,175444
Spinach, frozen, boiled, ½ cup57311,458229
Carrots, raw, ½ cup4599,189184
Pumpkin pie, commercially prepared, 1 piece4883,743249
Cantaloupe, raw, ½ cup1352,70654
Peppers, sweet, red, raw, ½ cup1172,33247
Mangos, raw, 1 whole1122,24045
Black-eyed peas (cowpeas), boiled, 1 cup661,30526
Apricots, dried, sulfured, 10 halves631,26125
Broccoli, boiled, ½ cup601,20824
Ice cream, French vanilla, soft serve, 1 cup2781,01420
Cheese, ricotta, part skim, 1 cup26394519
Tomato juice, canned, ¾ cup4282116
Herring, Atlantic, pickled, 3 ounces21973115
Ready-to-eat cereal, fortified with 10% of the DV for vitamin A, ¾–1 cup (more heavily fortified cereals might provide more of the DV)127–14950010
Milk, fat-free or skim, with added vitamin A and vitamin D, 1 cup14950010
Baked beans, canned, plain or vegetarian, 1 cup132745
Egg, hard boiled, 1 large752605
Summer squash, all varieties, boiled, ½ cup101914
Salmon, sockeye, cooked, 3 ounces591764
Yogurt, plain, low fat, 1 cup321162
Pistachio nuts, dry roasted, 1 ounce4731
Tuna, light, canned in oil, drained solids, 3 ounces20651
Chicken, breast meat and skin, roasted, ½ breast5180

Footnote: *DV = Daily Value. DVs were developed by the FDA to help consumers compare the nutrient contents of products within the context of a total diet. The DV for vitamin A is 5,000 IU for adults and children age 4 and older. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 43]

Beta Carotene

Beta-carotene is a red-orange pigment found in plants and fruits, especially carrots and colorful vegetables. It is the yellow/orange pigment that gives vegetables and fruits their rich colors.

Carotene is an orange photosynthetic pigment important for photosynthesis. It is responsible for the orange color of the carrot and many other fruits and vegetables. It contributes to photosynthesis by transmitting the light energy it absorbs to chlorophyll. Chemically, carotene is a terpene. It is the dimer of retinol (vitamin A) and comes in two primary forms: alpha- and beta-carotene. Gamma-, delta- and epsilon-carotene also exist. Carotene can be stored in the liver and converted to vitamin A as needed.

Beta-carotene in itself is not an essential nutrient, but vitamin A is.

Beta-carotene is a carotenoid that is a precursor of vitamin A and the human body converts beta-carotene into vitamin A (retinol). We need vitamin A for healthy skin and mucus membranes, our immune system, and good eye health and vision.

Beta-carotene, like all carotenoids, is an antioxidant. An antioxidant is a substance that inhibits the oxidation of other molecules; it protects the body from free radicals.

Free radicals damage cells through oxidation. Eventually, the damage caused by free radicals can cause several chronic illnesses.

Several studies have shown that antioxidants through diet help people’s immune systems, protect against free radicals, and lower the risk of developing cancer and heart disease.

Some studies have suggested that those who consume at least four daily servings of beta-carotene rich fruits and/or vegetables have a lower risk of developing cancer or heart disease.

Beta-carotene may also slow down cognitive decline. Men who have been taking beta-carotene supplements for 15 or more years are considerably less likely to experience cognitive decline than other males, researchers from Harvard Medical School reported in Archives of Internal Medicine.

beta carotene rich foods

B-group vitamins

There is a recent comprehensive review of B vitamins and immunity 44. B vitamins are involved in intestinal immune regulation, thus contributing to gut barrier function. Folic acid (vitamin B9) deficiency in animals causes thymus and spleen atrophy, and decreases circulating T lymphocyte numbers. Spleen lymphocyte proliferation is also reduced but the phagocytic and bactericidal capacity of neutrophils appears unchanged. In contrast, vitamin B12 deficiency decreases phagocytic and bacterial killing capacity of neutrophils, while vitamin B6 deficiency causes thymus and spleen atrophy, low blood T lymphocyte numbers and impaired lymphocyte proliferation and T lymphocyte-mediated immune responses. Vitamins B6 and B12 and folate all support the activity of natural killer cells and CD8+ cytotoxic T lymphocytes, effects which would be important in antiviral defence. Patients with vitamin B12 deficiency had low blood numbers of CD8+ T lymphocytes and low natural killer cell activity 45. In a study in healthy older humans 46, a vitamin B6-deficient diet for 21 days resulted in a decreased percentage and total number of circulating lymphocytes, and a decrease in T and B lymphocyte proliferation and IL-2 production. Repletion over 21 days using vitamin B6 at levels below those recommended did not return immune function to starting values, while repletion at the recommended intake (22.5 µg/kg body weight per day, which would be 1.575 mg/day in a 70 kg individual) did 46. Providing excess vitamin B6 (33.75 µg/kg body weight per day, which would be 2.362 mg/day in a 70 kg individual) for 4 days caused a further increase in lymphocyte proliferation and IL-2 production.

Thiamin (Vitamin B1)

Thiamin (or thiamine) is one of the water-soluble B vitamins. It is also known as vitamin B1. Thiamin is naturally present in some foods, added to some food products, and available as a dietary supplement. This vitamin plays a critical role in energy metabolism and, therefore, in the growth, development, and function of cells 47.

Thiamin (also called vitamin B1) helps turn the food you eat into the energy you need. Thiamin is important for the growth, development, and function of the cells in your body.

Table 7. Selected Food Sources of Thiamine (vitamin B1)

FoodMilligrams
(mg) per
serving
Percent DV*
Breakfast cereals, fortified with 100% of the DV for thiamin, 1 serving1.5100
Rice, white, long grain, enriched, parboiled, ½ cup1.473
Egg noodles, enriched, cooked, 1 cup0.533
Pork chop, bone-in, broiled, 3 ounces0.427
Trout, cooked, dry heat, 3 ounces0.427
Black beans, boiled, ½ cup0.427
English muffin, plain, enriched, 1 muffin0.320
Mussels, blue, cooked, moist heat, 3 ounces0.320
Tuna, Bluefin, cooked, dry heat, 3 ounces0.213
Macaroni, whole wheat, cooked, 1 cup0.213
Acorn squash, cubed, baked, ½ cup0.213
Rice, brown, long grain, not enriched, cooked, ½ cup0.17
Bread, whole wheat, 1 slice0.17
Orange juice, prepared from concentrate, 1 cup0.17
Sunflower seeds, toasted, 1 ounce0.17
Beef steak, bottom round, trimmed of fat, braised, 3 ounces0.17
Yogurt, plain, low fat, 1 cup0.17
Oatmeal, regular and quick, unenriched, cooked with water, ½ cup0.17
Corn, yellow, boiled, 1 medium ear0.17
Milk, 2%, 1 cup0.17
Barley, pearled, cooked, 1 cup0.17
Cheddar cheese, 1½ ounces00
Chicken, meat and skin, roasted, 3 ounces00
Apple, sliced, 1 cup00

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for thiamine is 1.5 mg for adults and children age 4 and older. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 48]

Vitamin B2 (Riboflavin)

Riboflavin also called vitamin B2 is one of the B vitamins, which are all water soluble and it’s important for the growth, development, and function of the cells in your body. It also helps turn the food you eat into the energy you need.

More than 90% of dietary riboflavin is in the form of flavin adenine dinucleotide (FAD) or flavin mononucleotide (FMN); the remaining 10% is comprised of the free form and glycosides or esters 49, 50. Most riboflavin is absorbed in the proximal small intestine 51. The body absorbs little riboflavin from single doses beyond 27 mg and stores only small amounts of riboflavin in the liver, heart, and kidneys. When excess amounts are consumed, they are either not absorbed or the small amount that is absorbed is excreted in urine 50.

Bacteria in the large intestine produce free riboflavin that can be absorbed by the large intestine in amounts that depend on the diet. More riboflavin is produced after ingestion of vegetable-based than meat-based foods 49.

Riboflavin is yellow and naturally fluorescent when exposed to ultraviolet light 52. Moreover, ultraviolet and visible light can rapidly inactivate riboflavin and its derivatives. Because of this sensitivity, lengthy light therapy to treat jaundice in newborns or skin disorders can lead to riboflavin deficiency. The risk of riboflavin loss from exposure to light is the reason why milk is not typically stored in glass containers 50, 53.

Table 8. Selected Food Sources of Riboflavin (vitamin B2)

FoodMilligrams (mg) per servingPercent DV*
Beef liver, pan fried, 3 ounces2.9171
Breakfast cereals, fortified with 100% of the DV for riboflavin, 1 serving1.7100
Oats, instant, fortified, cooked with water, 1 cup1.165
Yogurt, plain, fat free, 1 cup0.635
Milk, 2% fat, 1 cup0.529
Beef, tenderloin steak, boneless, trimmed of fat, grilled, 3 ounces0.424
Clams, mixed species, cooked, moist heat, 3 ounces0.424
Mushrooms, portabella, sliced, grilled, ½ cup0.318
Almonds, dry roasted, 1 ounce0.318
Cheese, Swiss, 3 ounces0.318
Rotisserie chicken, breast meat only, 3 ounces0.212
Egg, whole, scrambled, 1 large0.212
Quinoa, cooked, 1 cup0.212
Bagel, plain, enriched, 1 medium (3½”–4” diameter)0.212
Salmon, pink, canned, 3 ounces0.212
Spinach, raw, 1 cup0.16
Apple, with skin, 1 large0.16
Kidney beans, canned, 1 cup0.16
Macaroni, elbow shaped, whole wheat, cooked, 1 cup0.16
Bread, whole wheat, 1 slice0.16
Cod, Atlantic, cooked, dry heat, 3 ounces0.16
Sunflower seeds, toasted, 1 ounce0.16
Tomatoes, crushed, canned, ½ cup0.16
Rice, white, enriched, long grain, cooked, ½ cup0.16
Rice, brown, long grain, cooked, ½ cup00

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for riboflavin is 1.7 mg for adults and children age 4 and older. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 48]

Vitamin B6 (Pyridoxine)

Vitamin B6 includes a group of closely related compounds: pyridoxine, pyridoxal, and pyridoxamine. Substantial proportions of the naturally occurring pyridoxine in fruits, vegetables, and grains exist in glycosylated forms that exhibit reduced bioavailability 54. The body needs vitamin B6 for more than 100 enzyme reactions involved in metabolism. They are metabolized in the body to pyridoxal phosphate, which acts as a coenzyme in many important reactions in blood, CNS, and skin metabolism. Vitamin B6 is important in heme and nucleic acid biosynthesis and in lipid, carbohydrate, and amino acid metabolism. Vitamin B6 is also involved in brain development during pregnancy and infancy as well as immune function.

Vitamin B6 in coenzyme forms performs a wide variety of functions in the body and is extremely versatile, with involvement in more than 100 enzyme reactions, mostly concerned with protein metabolism. Both pyridoxal 5’ phosphate and pyridoxamine 5’ phosphate are involved in amino acid metabolism, and pyridoxal 5’ phosphate is also involved in the metabolism of one-carbon units, carbohydrates, and lipids 54. Vitamin B6 also plays a role in cognitive development through the biosynthesis of neurotransmitters and in maintaining normal levels of homocysteine, an amino acid in the blood 54. Vitamin B6 is involved in gluconeogenesis and glycogenolysis, immune function (for example, it promotes lymphocyte and interleukin-2 production), and hemoglobin formation 54.

The human body absorbs vitamin B6 in the jejunum. Phosphorylated forms of the vitamin are dephosphorylated, and the pool of free vitamin B6 is absorbed by passive diffusion 55.

Table 9. Selected Food Sources of Vitamin B6 (Pyridoxine)

FoodMilligrams (mg) per servingPercent DV*
Chickpeas, canned, 1 cup1.155
Beef liver, pan fried, 3 ounces0.945
Tuna, yellowfin, fresh, cooked, 3 ounces0.945
Salmon, sockeye, cooked, 3 ounces0.630
Chicken breast, roasted, 3 ounces0.525
Breakfast cereals, fortified with 25% of the DV for vitamin B60.525
Potatoes, boiled, 1 cup0.420
Turkey, meat only, roasted, 3 ounces0.420
Banana, 1 medium0.420
Marinara (spaghetti) sauce, ready to serve, 1 cup0.420
Ground beef, patty, 85% lean, broiled, 3 ounces0.315
Waffles, plain, ready to heat, toasted, 1 waffle0.315
Bulgur, cooked, 1 cup0.210
Cottage cheese, 1% low-fat, 1 cup0.210
Squash, winter, baked, ½ cup0.210
Rice, white, long-grain, enriched, cooked, 1 cup0.15
Nuts, mixed, dry-roasted, 1 ounce0.15
Raisins, seedless, ½ cup0.15
Onions, chopped, ½ cup0.15
Spinach, frozen, chopped, boiled, ½ cup0.15
Tofu, raw, firm, prepared with calcium sulfate, ½ cup0.15
Watermelon, raw, 1 cup0.15

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for vitamin B6 is 2 mg for adults and children age 4 and older. However, the FDA does not require food labels to list vitamin B6 content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 48]

Vitamin B12 (Cyanocobalamin)

Vitamin B12 is also known as Cyanocobalamin is a nutrient that helps keep the body’s nerve and blood cells healthy and helps make DNA, the genetic material in all cells. Vitamin B12 also helps prevent a type of anemia called megaloblastic anemia that makes people tired and weak.

Vitamin B12 is a water-soluble vitamin that is naturally present in some foods, added to others, and available as a dietary supplement and a prescription medication. Vitamin B12 exists in several forms and contains the mineral cobalt 56, 57, 58, 59, so compounds with vitamin B12 activity are collectively called “cobalamins”. Methylcobalamin and 5-deoxyadenosylcobalamin are the forms of vitamin B12 that are active in human metabolism 60.

Two steps are required for the body to absorb vitamin B12 from food.

  • First, food-bound vitamin B12 is released in the stomach’s acid environment (hydrochloric acid and and gastric protease in the stomach separate vitamin B12 from the protein to which vitamin B12 is attached in food) and is bound to R protein (haptocorrin) 60. When synthetic vitamin B12 is added to fortified foods and dietary supplements, it is already in free form and thus, does not require this separation step.
  • Second, pancreatic enzymes cleave this B12 complex (B12-R protein) in the small intestine. After cleavage, intrinsic factor (a protein made by the stomach), secreted by parietal cells in the gastric mucosa, binds with the free vitamin B12. Intrinsic factor is required for absorption of vitamin B12, which takes place in the terminal ileum 60, 61. Approximately 56% of a 1 mcg oral dose of vitamin B12 is absorbed, but absorption decreases drastically when the capacity of intrinsic factor is exceeded (at 1–2 mcg of vitamin B12) 62. Some people have pernicious anemia, a condition where they cannot make intrinsic factor. As a result, they have trouble absorbing vitamin B12 from all foods and dietary supplements.

Several food sources of vitamin B12 are listed in Table 10.

Table 10. Selected Food Sources of Vitamin B12

FoodMicrograms (mcg) per servingPercent DV*
Clams, cooked, 3 ounces84.11402
Liver, beef, cooked, 3 ounces70.71178
Breakfast cereals, fortified with 100% of the DV for vitamin B12, 1 serving6100
Trout, rainbow, wild, cooked, 3 ounces5.490
Salmon, sockeye, cooked, 3 ounces4.880
Trout, rainbow, farmed, cooked, 3 ounces3.558
Tuna fish, light, canned in water, 3 ounces2.542
Cheeseburger, double patty and bun, 1 sandwich2.135
Haddock, cooked, 3 ounces1.830
Breakfast cereals, fortified with 25% of the DV for vitamin B12, 1 serving1.525
Beef, top sirloin, broiled, 3 ounces1.423
Milk, low-fat, 1 cup1.218
Yogurt, fruit, low-fat, 8 ounces1.118
Cheese, Swiss, 1 ounce0.915
Beef taco, 1 soft taco0.915
Ham, cured, roasted, 3 ounces0.610
Egg, whole, hard boiled, 1 large0.610
Chicken, breast meat, roasted, 3 ounces0.35

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers determine the level of various nutrients in a standard serving of food in relation to their approximate requirement for it. The DV for vitamin B12 is 6.0 mcg. However, the FDA does not require food labels to list vitamin B12 content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient, but foods providing lower percentages of the DV also contribute to a healthful diet.

[Source 48]

Folate (Vitamin B9)

Folate is also known vitamin B9 (Folacin, Folic Acid, Pteroylglutamic acid) that is naturally present in many foods. Folic Acid is a form of folate that is manufactured and used in dietary supplements and fortified foods 63. Everyone needs folic acid. Our bodies need folate to make DNA and other genetic material. Folate is also needed for the body’s cells to divide.

Folic acid and folate also help your body make healthy new red blood cells. Red blood cells carry oxygen to all the parts of your body. If your body does not make enough red blood cells, you can develop anemia. Anemia happens when your blood cannot carry enough oxygen to your body, which makes you pale, tired, or weak. Also, if you do not get enough folic acid, you could develop a type of anemia called folate-deficiency anemia 64.

Folate-deficiency anemia is most common during pregnancy. Other causes of folate-deficiency anemia include alcoholism and certain medicines to treat seizures, anxiety, or arthritis.

In women and pregnant mothers, folic acid is very important because it can help prevent some major birth defects of the baby’s brain and spine (anencephaly and spina bifida) 65.

Every woman needs folic acid every day, whether she’s planning to get pregnant or not, for the healthy new cells the body makes daily. Think about the skin, hair, and nails. These – and other parts of the body – make new cells each day.

Centers for Disease Control and Prevention (CDC) urges women to take 400 mcg of folic acid every day, starting at least one month before getting pregnant and while she is pregnant, to help prevent major birth defects of the baby’s brain and spine.

Folate is naturally present in many foods and food companies add folic acid to other foods, including bread, cereal, and pasta. You can get recommended amounts by eating a variety of foods, including the following:

  • Leafy Green Vegetables (especially asparagus, Brussels sprouts, and dark green leafy vegetables such as spinach and mustard greens).
  • Fruits and fruit juices (especially oranges and orange juice).
  • Nuts, beans, and peas (such as peanuts, black-eyed peas, and kidney beans).
  • Grains (including whole grains; fortified cold cereals; enriched flour products such as bread, bagels, cornmeal, and pasta; and rice).
  • Folic acid is added to many grain-based products, enriched breads, cereals and corn masa flour (used to make corn tortillas and tamales, for example). To find out whether folic acid has been added to a food, check the product label.

Beef liver is high in folate but is also high in cholesterol, so limit the amount you eat. Only small amounts of folate are found in other animal foods like meats, poultry, seafood, eggs, and dairy products.

Table 11. Selected Food Sources of Folate and Folic Acid

Foodmcg DFE per servingPercent DV*
Beef liver, braised, 3 ounces21554
Spinach, boiled, ½ cup13133
Black-eyed peas (cowpeas), boiled, ½ cup10526
Breakfast cereals, fortified with 25% of the DV†10025
Rice, white, medium-grain, cooked, ½ cup†9023
Asparagus, boiled, 4 spears8922
Spaghetti, cooked, enriched, ½ cup†8321
Brussels sprouts, frozen, boiled, ½ cup7820
Lettuce, romaine, shredded, 1 cup6416
Avocado, raw, sliced, ½ cup5915
Spinach, raw, 1 cup5815
Broccoli, chopped, frozen, cooked, ½ cup5213
Mustard greens, chopped, frozen, boiled, ½ cup5213
Green peas, frozen, boiled, ½ cup4712
Kidney beans, canned, ½ cup4612
Bread, white, 1 slice†4311
Peanuts, dry roasted, 1 ounce4110
Wheat germ, 2 tablespoons4010
Tomato juice, canned, ¾ cup369
Crab, Dungeness, 3 ounces369
Orange juice, ¾ cup359
Turnip greens, frozen, boiled, ½ cup328
Orange, fresh, 1 small297
Papaya, raw, cubed, ½ cup277
Banana, 1 medium246
Yeast, baker’s, ¼ teaspoon236
Egg, whole, hard-boiled, 1 large226
Vegetarian baked beans, canned, ½ cup154
Cantaloupe, raw, 1 wedge144
Fish, halibut, cooked, 3 ounces123
Milk, 1% fat, 1 cup123
Ground beef, 85% lean, cooked, 3 ounces72
Chicken breast, roasted, ½ breast31

Footnote: * DV = Daily Value. The FDA developed DVs to help consumers compare the nutrient contents of products within the context of a total diet. The DV for folate is 400 mcg for adults and children aged 4 and older. However, the FDA does not require food labels to list folate content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

† Fortified with folic acid as part of the folate fortification program.

[Source 48]

Vitamin C

Vitamin C also known as ascorbic acid or ascorbate, is a water-soluble vitamin that is naturally present in some foods, added to others, and available as a dietary supplement. Vitamin C is synthesized from D-glucose or D-galactose by many plants and animals. However, humans lack the enzyme L-gulonolactone oxidase required for ascorbic acid synthesis and must obtain vitamin C through food or supplements 66, 67. Vitamin C is found in many fruits and vegetables, including citrus fruits, tomatoes, potatoes, red and green peppers, kiwifruit, broccoli, strawberries, brussels sprouts, and cantaloupe. In the body, vitamin C acts as an antioxidant, helping to protect cells from the damage caused by free radicals. Free radicals are compounds formed when our bodies convert the food we eat into energy. People are also exposed to free radicals in the environment from cigarette smoke, air pollution, and ultraviolet light from the sun.

The Recommended Dietary Allowance (RDA; average daily level of intake sufficient to meet the nutrient requirement of 97–98% healthy individuals) for vitamin C ranges from 15 to 115 mg for infants and children (depending on age) and from 75 to 120 mg for nonsmoking adults; people who smoke need 35 mg more per day 68. The intestinal absorption of vitamin C is regulated by at least one specific dose-dependent, active transporter 69. Cells accumulate vitamin C via a second specific transport protein. In vitro studies have found that oxidized vitamin C, or dehydroascorbic acid, enters cells via some facilitated glucose transporters and is then reduced internally to ascorbic acid. The physiologic importance of dehydroascorbic acid uptake and its contribution to overall vitamin C economy is unknown.

Vitamin C plays a role in collagen, carnitine, hormone, and amino acid formation. It is essential for wound healing and facilitates recovery from burns. Vitamin C is also an antioxidant, supports immune function, and facilitates the absorption of iron 70. Vitamin C also plays an important role in both innate and adaptive immunity, probably because of its antioxidant effects, antimicrobial and antiviral actions, and effects on immune system modulators 71. Vitamin C helps maintain epithelial integrity, enhance the differentiation and proliferation of B cells and T cells, enhance phagocytosis, normalize cytokine production, and decrease histamine levels 72. Vitamin C might also inhibit viral replication 73.

Vitamin C deficiency impairs immune function and increases susceptibility to infections 72. Some research suggests that supplemental vitamin C enhances immune function 74, but its effects might vary depending on an individual’s vitamin C status 75.

Vitamin C deficiency is uncommon in the United States, affecting only about 7% of individuals aged 6 years and older 76. People who smoke and those whose diets include a limited variety of foods (such as some older adults and people with alcohol or drug use disorders) are more likely than others to obtain insufficient amounts of vitamin C 74.

High-Dose vitamin C, when taken by intravenous (IV) infusion, vitamin C can reach much higher levels in the blood than when it is taken by mouth. Studies suggest that these higher levels of vitamin C may cause the death of cancer cells in the laboratory. Surveys of healthcare practitioners at United States complementary and alternative medicine conferences in recent years have shown that high-dose IV vitamin C is frequently given to patients as a treatment for infections, fatigue, and cancers, including breast cancer 77.

There are reviews of the role of vitamin C in immunity and in host susceptibility to infection 78. Vitamin C is required for collagen biosynthesis and is vital for maintaining epithelial integrity. Vitamin C also has roles in several aspects of immunity, including leucocyte migration to sites of infection, phagocytosis and bacterial killing, natural killer cell activity, T lymphocyte function (especially of CD8+ cytotoxic T lymphocytes) and antibody production. Jacob et al 79 showed that a vitamin C-deficient diet in healthy young adult humans decreased mononuclear cell vitamin C content by 50% and decreased the T lymphocyte-mediated immune responses to recall antigens. Vitamin C deficiency in animal models increases susceptibility to a variety of infections 78. People deficient in vitamin C are susceptible to severe respiratory infections such as pneumonia. A meta-analysis 24 reported a significant reduction in the risk of pneumonia with vitamin C supplementation, particularly in individuals with low dietary intakes. Vitamin C supplementation has also been shown to decrease the duration and severity of upper respiratory tract infections, such as the common cold, especially in people under enhanced physical stress 25.

Vitamin C is required for the biosynthesis of collagen, L-carnitine, and certain neurotransmitters; vitamin C is also involved in protein metabolism 67, 80. Collagen is an essential component of connective tissue, which plays a vital role in wound healing. Vitamin C is also an important physiological antioxidant 81 and has been shown to regenerate other antioxidants within the body, including alpha-tocopherol (vitamin E) 82. Ongoing research is examining whether vitamin C, by limiting the damaging effects of free radicals through its antioxidant activity, might help prevent or delay the development of certain cancers, cardiovascular disease, and other diseases in which oxidative stress plays a causal role. In addition to its biosynthetic and antioxidant functions, vitamin C plays an important role in immune function 82 and improves the absorption of nonheme iron 83, the form of iron present in plant-based foods. Insufficient vitamin C intake causes scurvy, which is characterized by fatigue or lassitude, widespread connective tissue weakness, and capillary fragility 67, 80, 82, 84, 85, 86, 87.

Table 12. Selected Food Sources of Vitamin C

FoodMilligrams (mg) per servingPercent (%) DV*
Red pepper, sweet, raw, ½ cup95158
Orange juice, ¾ cup93155
Orange, 1 medium70117
Grapefruit juice, ¾ cup70117
Kiwifruit, 1 medium64107
Green pepper, sweet, raw, ½ cup60100
Broccoli, cooked, ½ cup5185
Strawberries, fresh, sliced, ½ cup4982
Brussels sprouts, cooked, ½ cup4880
Grapefruit, ½ medium3965
Broccoli, raw, ½ cup3965
Tomato juice, ¾ cup3355
Cantaloupe, ½ cup2948
Cabbage, cooked, ½ cup2847
Cauliflower, raw, ½ cup2643
Potato, baked, 1 medium1728
Tomato, raw, 1 medium1728
Spinach, cooked, ½ cup915
Green peas, frozen, cooked, ½ cup813

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for vitamin C is 60 mg for adults and children aged 4 and older. The FDA requires all food labels to list the percent DV for vitamin C. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 48]

Vitamin D

Vitamin D is a fat-soluble vitamin that is naturally present in very few foods, added to others, and available as a dietary supplement. It is also produced endogenously when ultraviolet rays from sunlight strike the skin and trigger vitamin D synthesis. Vitamin D obtained from sun exposure, food, and supplements is biologically inert and must undergo two hydroxylations in the body for activation 88. The first occurs in the liver and converts vitamin D to 25-hydroxyvitamin D [25(OH)D], also known as calcidiol. The second occurs primarily in the kidney and forms the physiologically active 1,25-dihydroxyvitamin D [1,25(OH)2D], also known as calcitriol 89.

Vitamin D is a nutrient found in some foods that is needed for health and to maintain strong bones. It does so by helping the body absorb calcium (one of bone’s main building blocks) from food and supplements. People who get too little vitamin D may develop soft, thin, and brittle bones, a condition known as rickets in children and osteomalacia in adults.

Vitamin D is important to the body in many other ways as well. Muscles need it to move, for example, nerves need it to carry messages between the brain and every body part, and the immune system needs vitamin D to fight off invading bacteria and viruses. Together with calcium, vitamin D also helps protect older adults from osteoporosis. Vitamin D is found in cells throughout the body.

Vitamin D promotes calcium absorption in the gut and maintains adequate serum calcium and phosphate concentrations to enable normal mineralization of bone and to prevent hypocalcemic tetany. It is also needed for bone growth and bone remodeling by osteoblasts and osteoclasts 89, 90. Without sufficient vitamin D, bones can become thin, brittle, or misshapen. Vitamin D sufficiency prevents rickets in children and osteomalacia in adults 89. Together with calcium, vitamin D also helps protect older adults from osteoporosis.

Vitamin D has other roles in the body, including modulation of cell growth, neuromuscular and immune function, and reduction of inflammation 89, 91, 92. Many genes encoding proteins that regulate cell proliferation, differentiation, and apoptosis are modulated in part by vitamin D 89. Many cells have vitamin D receptors, and some convert 25(OH)D to 1,25(OH)2D.

There are a number of reviews of the role of vitamin D and its metabolites in immunity and in host susceptibility to infection 93. The active form of vitamin D (1,25-dihydroxyvitamin D3) is referred to here as vitamin D. Vitamin D receptors have been identified in most immune cells and some cells of the immune system can synthesise the active form of vitamin D from its precursor, suggesting that vitamin D is likely to have important immunoregulatory properties. Vitamin D enhances epithelial integrity and induces antimicrobial peptide (eg, cathelicidin) synthesis in epithelial cells and macrophages, directly enhancing host defence 94. However, the effects of vitamin D on the cellular components of immunity are rather complex. Vitamin D promotes differentiation of monocytes to macrophages and increases phagocytosis, superoxide production and bacterial killing by innate immune cells. It also promotes antigen processing by dendritic cells although antigen presentation may be impaired. Vitamin D is also reported to inhibit T-cell proliferation and production of cytokines by T helper 1 lymphocytes and of antibodies by B lymphocytes, highlighting the paradoxical nature of its effects. Effects on T helper 2 responses are not clear and vitamin D seems to increase number of regulatory T lymphocytes. Vitamin D seems to have little impact on CD8+ T lymphocytes. A systematic review and meta-analysis of the influence of vitamin D status on influenza vaccination (nine studies involving 2367 individuals) found lower seroprotection rates to influenza A virus subtype H3N2 and to influenza B virus in those who were vitamin D deficient 95. Berry et al 96 described an inverse linear relationship between vitamin D levels and respiratory tract infections in a cross-sectional study of 6789 British adults. In agreement with this, data from the US Third National Health and Nutrition Examination Survey which included 18 883 adults showed an independent inverse association between serum 25(OH)-vitamin D and recent upper respiratory tract infection 97. Other studies also report that individuals with low vitamin D status have a higher risk of viral respiratory tract infections 98. Supplementation of Japanese schoolchildren with vitamin D for 4 months during winter decreased the risk of influenza by about 40% 99. Meta-analyses have concluded that vitamin D supplementation can reduce the risk of respiratory tract infections 29, 28.

Table 13. Selected Food Sources of Vitamin D

FoodIUs per serving*Percent DV**
Cod liver oil, 1 tablespoon1360340
Swordfish, cooked, 3 ounces566142
Salmon (sockeye), cooked, 3 ounces447112
Tuna fish, canned in water, drained, 3 ounces15439
Orange juice fortified with vitamin D, 1 cup (check product labels, as amount of added vitamin D varies)13734
Milk, nonfat, reduced fat, and whole, vitamin D-fortified, 1 cup115-12429-31
Yogurt, fortified with 20% of the DV for vitamin D, 6 ounces (more heavily fortified yogurts provide more of the DV)8020
Margarine, fortified, 1 tablespoon6015
Sardines, canned in oil, drained, 2 sardines4612
Liver, beef, cooked, 3 ounces4211
Egg, 1 large (vitamin D is found in yolk)4110
Ready-to-eat cereal, fortified with 10% of the DV for vitamin D, 0.75-1 cup (more heavily fortified cereals might provide more of the DV)4010
Cheese, Swiss, 1 ounce62

Footnotes: * IUs = International Units. ** DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration to help consumers compare the nutrient contents among products within the context of a total daily diet. The DV for vitamin D is currently set at 400 IU for adults and children age 4 and older. Food labels, however, are not required to list vitamin D content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient, but foods providing lower percentages of the DV also contribute to a healthful diet.

[Source 48]

Vitamin E

Naturally occurring vitamin E exists in eight chemical forms (alpha-, beta-, gamma-, and delta-tocopherol and alpha-, beta-, gamma-, and delta-tocotrienol) that have varying levels of biological activity 100. Alpha- (or α-) tocopherol is the only form that is recognized to meet human requirements, but beta-, gamma-, and delta-tocopherols, 4 tocotrienols, and several stereoisomers may also have important biologic activity. These compounds act as antioxidants, which prevent lipid peroxidation of polyunsaturated fatty acids in cellular membranes 101.

Serum concentrations of vitamin E (alpha-tocopherol) depend on the liver, which takes up the nutrient after the various forms are absorbed from the small intestine.

Vitamin E is a fat-soluble antioxidant that stops the production of reactive oxygen species formed when fat undergoes oxidation. Scientists are investigating whether, by limiting free-radical production and possibly through other mechanisms, vitamin E might help prevent or delay the chronic diseases associated with free radicals.

Antioxidants protect cells from the damaging effects of free radicals, which are molecules that contain an unshared electron. Free radicals damage cells and might contribute to the development of cardiovascular disease and cancer 102. Unshared electrons are highly energetic and react rapidly with oxygen to form reactive oxygen species. The body forms reactive oxygen species endogenously when it converts food to energy, and antioxidants might protect cells from the damaging effects of reactive oxygen species. The body is also exposed to free radicals from environmental exposures, such as cigarette smoke, air pollution, and ultraviolet radiation from the sun. Reactive oxygen species are part of signaling mechanisms among cells.

The body also needs vitamin E to boost its immune system so that it can fight off invading bacteria and viruses. It helps to widen blood vessels and keep blood from clotting within them.

In addition to its activities as an antioxidant, vitamin E is involved in immune function and, as shown primarily by in vitro studies of cells, cell signaling, regulation of gene expression, and other metabolic processes 100. Alpha-tocopherol inhibits the activity of protein kinase C, an enzyme involved in cell proliferation and differentiation in smooth muscle cells, platelets, and monocytes 103. Vitamin-E–replete endothelial cells lining the interior surface of blood vessels are better able to resist blood-cell components adhering to this surface. Vitamin E also increases the expression of two enzymes that suppress arachidonic acid metabolism, thereby increasing the release of prostacyclin from the endothelium, which, in turn, dilates blood vessels and inhibits platelet aggregation 103.

There are a number of reviews of the role of vitamin E in immunity and host susceptibility to infection 104. In laboratory animals, vitamin E deficiency decreases lymphocyte proliferation, natural killer cell activity, specific antibody production following vaccination and phagocytosis by neutrophils. Vitamin E deficiency also increases susceptibility of animals to infectious pathogens. Vitamin E supplementation of the diet of laboratory animals enhances antibody production, lymphocyte proliferation, T helper 1-type cytokine production, natural killer cell activity and macrophage phagocytosis. Vitamin E promotes interaction between dendritic cells and CD4+ T lymphocytes. There is a positive association between plasma vitamin E and cell-mediated immune responses, and a negative association has been demonstrated between plasma vitamin E and the risk of infections in healthy adults over 60 years of age 105. There appears to be particular benefit of vitamin E supplementation for the elderly 106. Studies by Meydani et al 107 demonstrated that vitamin E supplementation at high doses, one study used 800 mg/day 108 and the other used doses of 60, 200 and 800 mg/day 107 enhanced T helper 1 cell-mediated immunity (lymphocyte proliferation, IL-2 production) and improved vaccination responses, including to hepatitis B virus. Supplementation of older adults with vitamin E (200 mg/day) improved neutrophil chemotaxis and phagocytosis, natural killer cell activity and mitogen-induced lymphocyte proliferation 106. Secondary analysis of data from the Alpha-Tocopherol, Beta Carotene Cancer Prevention Study identified that daily vitamin E supplements for 5 to 8 years reduced the incidence of hospital treated, community-acquired pneumonia in smokers 109. One study reported that vitamin E supplementation (200 IU/day~135 mg/day) for 1 year decreased risk of upper respiratory tract infections in the elderly 110, but another study did not see an effect of supplemental vitamin E (200 mg/day) on the incidence, duration or severity of respiratory infections in an elderly population 111.

Table 14. Selected Food Sources of Vitamin E (Alpha-Tocopherol)

FoodMilligrams (mg) per servingPercent DV*
Wheat germ oil, 1 tablespoon20.3100
Sunflower seeds, dry roasted, 1 ounce7.437
Almonds, dry roasted, 1 ounce6.834
Sunflower oil, 1 tablespoon5.628
Safflower oil, 1 tablespoon4.625
Hazelnuts, dry roasted, 1 ounce4.322
Peanut butter, 2 tablespoons2.915
Peanuts, dry roasted, 1 ounce2.211
Corn oil, 1 tablespoon1.910
Spinach, boiled, ½ cup1.910
Broccoli, chopped, boiled, ½ cup1.26
Soybean oil, 1 tablespoon1.16
Kiwifruit, 1 medium1.16
Mango, sliced, ½ cup0.74
Tomato, raw, 1 medium0.74
Spinach, raw, 1 cup0.63

Footnote: *DV = Daily Value. DVs were developed by the FDA to help consumers compare the nutrient content of different foods within the context of a total diet. The DV for vitamin E is 30 IU (approximately 20 mg of natural alpha-tocopherol) for adults and children age 4 and older. However, the FDA does not require food labels to list vitamin E content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient, but foods providing lower percentages of the DV also contribute to a healthful diet.

[Source 48]

Iron

Iron is a mineral that our bodies need for many functions. In the human body, iron is present in all cells and has several vital functions — as a carrier of oxygen to the tissues from the lungs in the form of hemoglobin (Hb), as a facilitator of oxygen use and storage in the muscles as myoglobin, as a transport medium for electrons within the cells in the form of cytochromes, and as an integral part of enzyme reactions in various tissues. Too little iron can interfere with these vital functions and lead to morbidity and mortality 112, 113.

In adults, the recommended dietary allowance of iron is 8 to 11 mg per day for men and 8 to 18 mg for women in whom higher levels are recommended during pregnancy (27 mg per day) 114. Iron is poorly absorbed and body and tissue iron stores are controlled largely by modifying rates of absorption. Adequate amounts of iron are found in most Western diets, with highest levels found in red meats and moderate levels in fish, poultry, green vegetables, cereals and grains (some of which are fortified with iron).

Your body needs the right amount of iron. If you have too little iron, you may develop iron deficiency anemia. Iron deficiency is usually due to loss of iron, predominantly as a result of blood loss in the gastrointestinal tract or from menstruation and is rarely due to deficiency in intake or an inability to absorb enough iron from foods. People at higher risk of having too little iron are young children and women who are pregnant or have periods.

There are a number of reviews of the role of iron in immunity and host susceptibility to infection 115. Iron deficiency induces thymus atrophy, reducing output of naive T lymphocytes, and has multiple effects on immune function in humans. The effects are wide ranging and include impairment of respiratory burst and bacterial killing, natural killer cell activity, T lymphocyte proliferation and production of T helper 1 cytokines. T lymphocyte proliferation was lower by 50% to 60% in iron-deficient than in iron-replete housebound older Canadian women 116. These observations would suggest a clear case for iron deficiency increasing susceptibility to infection. However, the relationship between iron deficiency and susceptibility to infection remains complex 117. Evidence suggests that infections caused by organisms that spend part of their life-cycle intracellularly, such as plasmodia and mycobacteria, may actually be enhanced by iron. In the tropics, in children of all ages, iron at doses above a particular threshold has been associated with increased risk of malaria and other infections, including pneumonia. Thus, iron intervention in malaria-endemic areas is not advised, particularly high doses in the young, those with compromised immunity and during the peak malaria transmission season. There are different explanations for the detrimental effects of iron administration on infections. First, iron overload causes impairment of immune function 118. Second, excess iron favors damaging inflammation. Third, micro-organisms require iron and providing it may favor the growth of the pathogen. Perhaps for the latter reasons several host immune mechanisms have developed for withholding iron from a pathogen 115. In a recent study giving iron (50 mg on each of 4 days a week) to iron-deficient school children in South Africa increased the risk of respiratory infections 119; coadministration of omega-3 fatty acids (500 mg on each of 4 days a week) mitigated the effect of iron. Meta-analysis of studies in Chinese children showed that those with recurrent respiratory tract infection were more likely to have low hair iron 30.

What foods provide iron?

Iron is found naturally in many foods and is added to some fortified food products. You can get recommended amounts of iron by eating a variety of foods, including the following:

  • Lean meat, seafood, and poultry.
  • Iron-fortified breakfast cereals and breads.
  • White beans, lentils, spinach, kidney beans, and peas.
  • Nuts and some dried fruits, such as raisins.

Iron in food comes in two forms: heme iron and nonheme iron. Nonheme iron is found in plant foods and iron-fortified food products. Meat, seafood, and poultry have both heme and nonheme iron.

Heme iron has higher bioavailability than nonheme iron, and other dietary components have less effect on the bioavailability of heme than nonheme iron 120. The bioavailability of iron is approximately 14% to 18% from mixed diets that include substantial amounts of meat, seafood, and vitamin C (ascorbic acid, which enhances the bioavailability of nonheme iron) and 5% to 12% from vegetarian diets 121. In addition to ascorbic acid, meat, poultry, and seafood can enhance nonheme iron absorption, whereas phytate (present in grains and beans) and certain polyphenols in some non-animal foods (such as cereals and legumes) have the opposite effect 122. Unlike other inhibitors of iron absorption, calcium might reduce the bioavailability of both nonheme and heme iron. However, the effects of enhancers and inhibitors of iron absorption are attenuated by a typical mixed western diet, so they have little effect on most people’s iron status.

Several food sources of iron are listed in Table 15. Some plant-based foods that are good sources of iron, such as spinach, have low iron bioavailability because they contain iron-absorption inhibitors, such as polyphenols 123.

Your body absorbs iron from plant sources better when you eat it with meat, poultry, seafood, and foods that contain vitamin C, like citrus fruits, strawberries, sweet peppers, tomatoes, and broccoli.

Table 15. Selected Food Sources of Iron

FoodMilligrams per servingPercent DV*
Breakfast cereals, fortified with 100% of the DV for iron, 1 serving18100
Oysters, eastern, cooked with moist heat, 3 ounces844
White beans, canned, 1 cup844
Chocolate, dark, 45%–69% cacao solids, 3 ounces739
Beef liver, pan fried, 3 ounces528
Lentils, boiled and drained, ½ cup317
Spinach, boiled and drained, ½ cup317
Tofu, firm, ½ cup317
Kidney beans, canned, ½ cup211
Sardines, Atlantic, canned in oil, drained solids with bone, 3 ounces211
Chickpeas, boiled and drained, ½ cup211
Tomatoes, canned, stewed, ½ cup211
Beef, braised bottom round, trimmed to 1/8” fat, 3 ounces211
Potato, baked, flesh and skin, 1 medium potato211
Cashew nuts, oil roasted, 1 ounce (18 nuts)211
Green peas, boiled, ½ cup16
Chicken, roasted, meat and skin, 3 ounces16
Rice, white, long grain, enriched, parboiled, drained, ½ cup16
Bread, whole wheat, 1 slice16
Bread, white, 1 slice16
Raisins, seedless, ¼ cup16
Spaghetti, whole wheat, cooked, 1 cup16
Tuna, light, canned in water, 3 ounces16
Turkey, roasted, breast meat and skin, 3 ounces16
Nuts, pistachio, dry roasted, 1 ounce (49 nuts)16
Broccoli, boiled and drained, ½ cup16
Egg, hard boiled, 1 large16
Rice, brown, long or medium grain, cooked, 1 cup16
Cheese, cheddar, 1.5 ounces00
Cantaloupe, diced, ½ cup00
Mushrooms, white, sliced and stir-fried, ½ cup00
Cheese, cottage, 2% milk fat, ½ cup00
Milk, 1 cup00

Footnote: * DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for iron is 18 mg for adults and children age 4 and older. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 48]

Copper

Copper is an essential mineral that you need to stay healthy. Your body uses copper to carry out many important functions, including making energy, connective tissues, and blood vessels. Copper also helps maintain the nervous, pigmentation, and immune systems, and activates genes. Your body also needs copper for brain development 124. In addition, defense against oxidative damage depends mainly on the copper-containing superoxide dismutases 125. Copper is a cofactor for several enzymes known as “cuproenzymes” involved in energy production, iron metabolism, neuropeptide activation, connective tissue synthesis, and neurotransmitter synthesis 124. One abundant cuproenzyme is ceruloplasmin, which plays a role in iron metabolism and carries more than 95% of the total copper in healthy human plasma 126.

There are a number of reviews of the role of copper in immunity and host susceptibility to infection 127. Copper itself has antimicrobial properties. Copper supports neutrophil, monocyte and macrophage function and natural killer cell activity. It promotes T lymphocyte responses such as proliferation and IL-2 production. Copper deficiency in animals impairs a range of immune functions and increases susceptibility to bacterial and parasitic challenges. Human studies show that subjects on a low copper diet have decreased lymphocyte proliferation and IL-2 production, with copper administration reversing these effects 128. Children with Menke’s syndrome, a rare congenital disease with complete absence of the circulating copper-carrying protein caeruloplasmin, show immune impairments and have increased bacterial infections, diarrhea and pneumonia 129. Meta-analysis of studies in Chinese children showed that those with recurrent respiratory tract infection were more likely to have low hair copper 30.

A wide variety of plant and animal foods contain copper, and the average human diet provides approximately 1,400 mcg/day for men and 1,100 mcg/day for women that is primarily absorbed in the upper small intestine 130. Almost two-thirds of the body’s copper is located in the skeleton and muscle 124.

Only small amounts of copper are typically stored in the body, and the average adult has a total body content of 50–120 mg copper 124. Most copper is excreted in bile, and a small amount is excreted in urine. Total fecal losses of copper of biliary origin and nonabsorbed dietary copper are about 1 mg/day 124. Copper levels in the body are homeostatically maintained by copper absorption from the intestine and copper release by the liver into bile to provide protection from copper deficiency and toxicity 131.

Copper status is not routinely assessed in clinical practice, and no biomarkers that accurately and reliably assess copper status have been identified 132. Human studies typically measure copper and cuproenzyme activity in plasma and blood cells because individuals with known copper deficiency often have low blood levels of copper and ceruloplasmin 132. However, plasma ceruloplasmin and copper levels can be influenced by other factors, such as estrogen status, pregnancy, infection, inflammation, and some cancers 132. Normal serum concentrations are 10–25 mcmol/L (63.5–158.9 mcg/dL) for copper and 180–400 mg/L for ceruloplasmin 133.

The amount of copper you need each day depends on your age. Typical diets in the United States meet or exceed the copper recommended dietary allowance (RDA), which is the average daily level of intake sufficient to meet the nutrient requirements of nearly all (97%–98%) healthy individuals; often used to plan nutritionally adequate diets for individuals. Mean dietary intakes of copper from foods range from 800 to 1,000 mcg per day for children aged 2–19 134. In adults aged 20 and older, average daily intakes of copper from food are 1,400 mcg for men and 1,100 mcg for women. Total intakes from supplements and foods are 900 to 1,100 mcg/day for children and 1,400 to 1,700 mcg/day for adults aged 20 and over.

Copper deficiency is uncommon in humans 132. Based on studies in animals and humans, the effects of copper deficiency include anemia, hypopigmentation, hypercholesterolemia, connective tissue disorders, osteoporosis and other bone defects, abnormal lipid metabolism, ataxia, and increased risk of infection 135.

Copper is available in dietary supplements containing only copper, in supplements containing copper in combination with other ingredients, and in many multivitamin/multimineral products 136. These supplements contain many different forms of copper, including cupric oxide, cupric sulfate, copper amino acid chelates, and copper gluconate. To date, no studies have compared the bioavailability of copper from these and other forms 137. The amount of copper in dietary supplements typically ranges from a few micrograms to 15 mg (about 17 times the daily value [DV] for copper) 136.

Table 16. Selected Food Sources of Copper

FoodMicrograms (mcg) per servingPercent
DV*
Beef, liver, pan fried (3 ounces)124001378
Oysters, eastern, wild, cooked, 3 ounces4850539
Baking chocolate, unsweetened, 1 ounce938104
Potatoes, cooked, flesh and skin, 1 medium potato67575
Mushrooms, shiitake, cooked, cut pieces, ½ cup65072
Cashew nuts, dry roasted, 1 ounce62970
Crab, Dungeness, cooked, 3 ounces62469
Sunflower seed kernels, toasted, ¼ cup61568
Turkey, giblets, simmered, 3 ounces58865
Chocolate, dark, 70%-85% cacao solids, 1 ounce50156
Tofu, raw, firm, ½ cup47653
Chickpeas, mature sees, ½ cup28932
Millet, cooked, 1 cup28031
Salmon, Atlantic, wild, cooked, 3 ounces27330
Pasta, whole wheat, cooked, 1 cup (not packed)26329
Avocado, raw, ½ cup21924
Figs, dried, ½ cup21424
Spinach, boiled, drained, ½ cup15717
Asparagus, cooked, drained, ½ cup14917
Seseame seeds, ¼ cup14716
Turkey, ground, cooked, 3 ounces12814
Cereals, Cream of Wheat, cooked with water, stove-top, 1 cup10412
Tomatoes, raw, chopped, ½ cup536
Yogurt, Greek, plain, lowfat, 7-ounce container425
Milk, nonfat, 1 cup273
Apples, raw, with skin, ½ cup slices172

Footnote: *DV = Daily Value. The U.S. Food and Drug Administration (FDA) developed DVs to help consumers compare the nutrient contents of foods and dietary supplements within the context of a total diet. The DV for copper is 0.9 mg (900 mcg) for adults and children age 4 years and older [13]. The FDA does not require food labels to list copper content unless copper has been added to the food. Foods providing 20% or more of the DV are considered to be high sources of a nutrient, but foods providing lower percentages of the DV also contribute to a healthful diet.

[Source 48]

Selenium

Selenium is a trace element that is naturally present in many foods, added to others, and available as a dietary supplement. Selenium, which is nutritionally essential for humans, is a constituent of more than two dozen selenoproteins that play critical roles in reproduction, thyroid hormone metabolism, DNA synthesis, and protection from oxidative damage and infection 138.

Selenium exists in two forms:

  • inorganic (selenate and selenite) and
  • organic (selenomethionine and selenocysteine) 139.

Both forms can be good dietary sources of selenium 140. Soils contain inorganic selenites and selenates that plants accumulate and convert to organic forms, mostly selenocysteine and selenomethionine and their methylated derivatives.

Selenium is found naturally in many foods. The amount of selenium in plant foods depends on the amount of selenium in the soil where they were grown. The amount of selenium in animal products depends on the selenium content of the foods that the animals ate. You can get recommended amounts of selenium by eating a variety of foods, including the following:

  • Seafood
  • Meat, poultry, eggs, and dairy products
  • Breads, cereals, and other grain products.

Selenium is important for reproduction, thyroid gland function, DNA production, and protecting the body from damage caused by free radicals and from infection 141, 142. Selenium is incorporated into selenoproteins that have a wide range of pleiotropic effects, ranging from antioxidant and anti-inflammatory effects to the production of active thyroid hormone 143. In the past 10 years, the discovery of disease-associated polymorphisms in selenoprotein genes has drawn attention to the relevance of selenoproteins to health. Low selenium status has been associated with increased risk of mortality, poor immune function, and cognitive decline. Higher selenium status or selenium supplementation has antiviral effects, is essential for successful male and female reproduction, and reduces the risk of autoimmune thyroid disease. Prospective studies have generally shown some benefit of higher selenium status on the risk of prostate, lung, colorectal, and bladder cancers, but findings from trials have been mixed, which probably emphasises the fact that supplementation will confer benefit only if intake of a nutrient is inadequate. Supplementation of people who already have adequate intake with additional selenium might increase their risk of type-2 diabetes. The crucial factor that needs to be emphasised with regard to the health effects of selenium is the inextricable U-shaped link with status; whereas additional selenium intake may benefit people with low status, those with adequate-to-high status might be affected adversely and should not take selenium supplements.

There are a number of reviews of the role of selenium in immunity and host susceptibility to infection 144. Selenium deficiency in laboratory animals adversely affects several components of both innate and acquired immunity, including T and B lymphocyte function including antibody production and increases susceptibility to infections. Lower selenium concentrations in humans have been linked with diminished natural killer cell activity and increased mycobacterial disease. Selenium deficiency was shown to permit mutations of coxsackievirus, polio virus and murine influenza virus increasing virulence 145. These latter observations suggest that poor selenium status could result in the emergence of more pathogenic strains of virus, thereby increasing the risks and burdens associated with viral infection. Selenium supplementation (100 to 300 µg/day depending on the study) has been shown to improve various aspects of immune function in humans 146, including in the elderly 147. Selenium supplementation (50 or 100 µg/day) in adults in the UK with low selenium status improved some aspects of their immune response to a poliovirus vaccine 148.

Table 17. Selected Food Sources of Selenium

FoodMicrograms (mcg) per servingPercent DV*
Brazil nuts, 1 ounce (6–8 nuts)544777
Tuna, yellowfin, cooked, dry heat, 3 ounces92131
Halibut, cooked, dry heat, 3 ounces4767
Sardines, canned in oil, drained solids with bone, 3 ounces4564
Ham, roasted, 3 ounces4260
Shrimp, canned, 3 ounces4057
Macaroni, enriched, cooked, 1 cup3753
Beef steak, bottom round, roasted, 3 ounces3347
Turkey, boneless, roasted, 3 ounces3144
Beef liver, pan fried, 3 ounces2840
Chicken, light meat, roasted, 3 ounces2231
Cottage cheese, 1% milkfat, 1 cup2029
Rice, brown, long-grain, cooked, 1 cup1927
Beef, ground, 25% fat, broiled, 3 ounces1826
Egg, hard-boiled, 1 large1521
Puffed wheat ready-to-eat cereal, fortified, 1 cup1521
Bread, whole-wheat, 1 slice1319
Baked beans, canned, plain or vegetarian, 1 cup1319
Oatmeal, regular and quick, unenriched, cooked with water, 1 cup1319
Spinach, frozen, boiled, 1 cup1116
Milk, 1% fat, 1 cup811
Yogurt, plain, low fat, 1 cup811
Lentils, boiled, 1 cup69
Bread, white, 1 slice69
Spaghetti sauce, marinara, 1 cup46
Cashew nuts, dry roasted, 1 ounce34
Corn flakes, 1 cup23
Green peas, frozen, boiled, 1 cup23
Bananas, sliced, 1 cup23
Potato, baked, flesh and skin, 1 potato11
Peaches, canned in water, solids and liquids, 1 cup11
Carrots, raw, 1 cup00
Lettuce, iceberg, raw, 1 cup00

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for selenium is 70 mcg for adults and children aged 4 and older. Foods providing 20% or more of the DV are considered to be high sources of a nutrient. The U.S. Department of Agriculture’s (USDA’s) Nutrient Database Web site 149 lists the nutrient content of many foods and provides a comprehensive list of foods containing selenium arranged by nutrient content and by food name.

[Source 48]

Zinc

Zinc is involved in numerous aspects of cellular metabolism. It is required for the catalytic activity of approximately 100 enzymes, including many nicotinamide adenine dinucleotide (NADH) dehydrogenases, RNA and DNA polymerases, and DNA transcription factors as well as alkaline phosphatase, superoxide dismutase, and carbonic anhydrase 150, 151 and it plays a role in immune function 152, 153, protein synthesis 153, wound healing 154, DNA synthesis 151, 153 and cell division 153. Zinc also supports normal growth and development during pregnancy, childhood, and adolescence 155, 156, 157 and is required for proper sense of taste and smell 158. A daily intake of zinc is required to maintain a steady state because the body has no specialized zinc storage system 159.

Most Americans get enough zinc from the foods they eat.

There are a number of reviews of the role of zinc in immunity and host susceptibility to infection 160. Of note, Read et al 161 have recently provided a very insightful evaluation of the role of zinc in antiviral immunity. Zinc inhibits the RNA polymerase required by RNA viruses, like coronaviruses, to replicate 162, suggesting that zinc may play a key role in host defence against RNA viruses. In vitro replication of influenza virus was inhibited by the zinc ionophore pyrrolidine dithiocarbamate 163 and there are indications that zinc might inhibit replication of SARS-associated coronavirus (SARS-CoV) in vitro 164. In addition, as discussed by Read et al 161, the zinc-binding metallothioneins seem to play an important role in antiviral defence 165. Zinc deficiency has a marked impact on bone marrow, decreasing the number immune precursor cells, with reduced output of naive B lymphocytes and causes thymic atrophy, reducing output of naive T lymphocytes. Therefore, zinc is important in maintaining T and B lymphocyte numbers. Zinc deficiency impairs many aspects of innate immunity, including phagocytosis, respiratory burst and natural killer cell activity. Zinc also supports the release of neutrophil extracellular traps that capture microbes 166. There are also marked effects of zinc deficiency on acquired immunity. Circulating CD4+ T lymphocyte numbers and function (eg, IL-2 and IFN-γ production) are decreased and there is a disturbance in favour of T helper 2 cells. Likewise, B lymphocyte numbers and antibody production are decreased in zinc deficiency. Zinc supports proliferation of CD8+ cytotoxic T lymphocytes, key cells in antiviral defence. Many of the in vitro immune effects of zinc are prevented by zinc chelation 167. Moderate or mild zinc deficiency or experimental zinc deficiency in humans result in decreased natural killer cell activity, T lymphocyte proliferation, IL-2 production and cell-mediated immune responses which can all be corrected by zinc repletion 168. In patients with zinc deficiency related to sickle cell disease, natural killer cell activity is decreased, but can be returned to normal by zinc supplementation 169. Patients with the zinc malabsorption syndrome acrodermatitis enteropathica display severe immune impairments 170 and increased susceptibility to bacterial, viral and fungal infections. Zinc supplementation (30 mg/day) increased T lymphocyte proliferation in elderly care home residents in the USA, an effect mainly due to an increase in numbers of T lymphocytes 171. The wide ranging impact of zinc deficiency on immune components is an important contributor to the increased susceptibility to infections, especially lower respiratory tract infection and diarrhoea, seen in zinc deficiency. Correcting zinc deficiency lowers the likelihood of diarrhea and of respiratory and skin infections, although some studies fail to show benefit of zinc supplementation in respiratory disease 172. Meta-analysis of studies in Chinese children showed that those with recurrent respiratory tract infection were more likely to have low hair zinc 30. Recent systematic reviews and meta-analyses of trials with zinc report shorter duration of common cold in adults 31, reduced incidence and prevalence of pneumonia in children 33 and reduced mortality when given to adults with severe pneumonia 34.

Table 18. Selected Food Sources of Zinc

FoodMilligrams (mg) per servingPercent DV*
Oysters, cooked, breaded and fried, 3 ounces74493
Beef chuck roast, braised, 3 ounces747
Crab, Alaska king, cooked, 3 ounces6.543
Beef patty, broiled, 3 ounces5.335
Breakfast cereal, fortified with 25% of the DV for zinc, ¾ cup serving3.825
Lobster, cooked, 3 ounces3.423
Pork chop, loin, cooked, 3 ounces2.919
Baked beans, canned, plain or vegetarian, ½ cup2.919
Chicken, dark meat, cooked, 3 ounces2.416
Yogurt, fruit, low fat, 8 ounces1.711
Cashews, dry roasted, 1 ounce1.611
Chickpeas, cooked, ½ cup1.39
Cheese, Swiss, 1 ounce1.28
Oatmeal, instant, plain, prepared with water, 1 packet1.17
Milk, low-fat or non fat, 1 cup17
Almonds, dry roasted, 1 ounce0.96
Kidney beans, cooked, ½ cup0.96
Chicken breast, roasted, skin removed, ½ breast0.96
Cheese, cheddar or mozzarella, 1 ounce0.96
Peas, green, frozen, cooked, ½ cup0.53
Flounder or sole, cooked, 3 ounces0.32

Footnote: * DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration to help consumers compare the nutrient contents of products within the context of a total diet. The DV for zinc is 15 mg for adults and children age 4 and older. Food labels, however, are not required to list zinc content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 48 ]

Magnesium

Magnesium (Mg) is an abundant mineral in your body that your body needs to stay healthy and is naturally present in many foods, added to other food products, available as a dietary supplement, and present in some medicines (such as antacids and laxatives) 173. Magnesium is important for many processes in your body, including regulating muscle and nerve function, energy production, blood sugar levels, and blood pressure and making protein, bone, and DNA. Magnesium is a cofactor in more than 300 enzyme systems that regulate diverse biochemical reactions in the body, including protein synthesis, muscle and nerve function, blood glucose control, and blood pressure regulation 174, 175, 176, 177, 178. Low magnesium levels (hypomagnesemia) don’t cause symptoms in the short term. However, chronically low magnesium levels can increase your risk of high blood pressure, heart disease, type 2 diabetes and osteoporosis 179.

Magnesium is required for energy production, oxidative phosphorylation, and glycolysis 180. It contributes to the structural development of bone and is required for the synthesis of DNA, RNA, and the antioxidant glutathione. Magnesium also plays a role in the active transport of calcium and potassium ions across cell membranes, a process that is important to nerve impulse conduction, muscle contraction, and normal heart rhythm 176. Many people don’t get enough magnesium in their diets. However, before you reach for a supplement, though, you should know that just a few servings of magnesium-rich foods a day can meet your need for this important nutrient.

An adult body contains approximately 25 g magnesium, with 50% to 60% present in the bones and most of the rest in soft tissues 181. Less than 1% of total magnesium is in blood serum, and these levels are kept under tight control through its absorption, reservoir, and excretion process by various organs such as the gut, bone, and kidney 182. Besides these organs, several hormones, namely vitamin D, parathyroid hormone, and estrogen, are involved in the regulation of normal level of magnesium 183. Normal serum magnesium concentrations range between 0.75 and 0.95 millimoles (mmol)/L 174, 184. Hypomagnesemia is defined as a serum magnesium level less than 0.75 mmol/L 185. Magnesium homeostasis is largely controlled by the kidney, which typically excretes about 120 mg magnesium into the urine each day 175. Urinary excretion is reduced when magnesium status is low 174. Hypomagnesemia is characterized by tetany (involuntary contraction of muscles), convulsion (seizures), and cardiac arrhythmia 186. Clinical and preclinical studies revealed that the magnesium level is found to be low in various pathological conditions such as migraine, diabetes, osteoporosis, asthma, preeclampsia, cardiovascular diseases and its correction is an important treatment strategy for these conditions 187, 188, 189, 190, 191.

The diets of many people in the United States provide less than the recommended amounts of magnesium. Men older than 70 and teenage girls and boys are most likely to have low intakes of magnesium. When the amount of magnesium people get from food and dietary supplements is combined, however, total intakes of magnesium are generally above recommended amounts.

Assessing magnesium status is difficult because most magnesium is inside cells or in bone 176. The most commonly used and readily available method for assessing magnesium status is measurement of serum magnesium concentration, even though serum levels have little correlation with total body magnesium levels or concentrations in specific tissues 185. Other methods for assessing magnesium status include measuring magnesium concentrations in erythrocytes, saliva, and urine; measuring ionized magnesium concentrations in blood, plasma, or serum; and conducting a magnesium-loading (or “tolerance”) test. No single method is considered satisfactory 192. Some experts 181 but not others 176 consider the tolerance test (in which urinary magnesium is measured after parenteral infusion of a dose of magnesium) to be the best method to assess magnesium status in adults. To comprehensively evaluate magnesium status, both laboratory tests and a clinical assessment might be required 185.

Magnesium is important in acquired immunity via regulating lymphocyte growth 193. An in vitro study (test tube study) carried out in chicken B cell line DT40 revealed that the removal of magnesium channel, TRPM7, results in cell death and can be partially corrected by magnesium supplementation 194. Mutation in a magnesium transporter, MagT1, is reported in patients with X-linked immunodeficiency diseases, Epstein–Barr virus infection, and neoplasia 195. Low CD4+ T cells and defective activation of T-lymphocytes are due to the decreased magnesium influx, which fails to activate PLCγ1 196. The importance of magnesium for CD4+ activation is also evident from reported studies conducted in asthma patients 197. However, further studies are essential to conclude the effect of magnesium on T cell signaling.

Magnesium has an important role in synthesizing and releasing immune cells and other associated processes like cell adhesion and phagocytosis 198. Magnesium acts as a natural calcium antagonist, the molecular basis for inflammatory response could be the result of modulation of intracellular calcium concentration 199. Besides, magnesium acts as a cofactor for the synthesis of immunoglobulin, CI 3 convertase, antibody-dependent cytolysis, macrophage responses to lymphocyte, IgM lymphocyte binding, T helper B cell adherence, substance P binding with lymphoblast, and binding of antigen to macrophage 200, 201. Magnesium deficiency affects various immune functions like the decline in NK cell level, monocytes and T cell ratio, increased oxidative stress after strenuous exercise, and elevated cytokine IL-6 level and inflammatory events. Deficiency of magnesium may be prone to recurrent bacterial and fungal infection 202. Many studies have demonstrated that in humans, a moderate or subclinical magnesium deficiency can induce chronic low-grade inflammation or exacerbate inflammatory stress caused by other factors 203. This low-grade inflammation increases the secretion of pro-inflammatory cytokines, which stimulate the resorption of bone by the induction of the differentiation of osteoclasts from their precursors 204. The ability of magnesium to decrease the inflammatory response and oxidative stress, as well as improving lung inflammation, possibly by inhibiting IL-6 pathway, NF-κB pathway, and L-type calcium channels 205, has raised the hypothesis of a possible magnesium supplementation in the prevention and treatment of COVID-19 patients, as suggested in the recent papers by Tang et al 206 and Iotti 207. Based Tang et al 208 basic and clinical research study, it is evident that magnesium effectively treats respiratory diseases like asthma and pneumonia because of its anti-inflammatory, antioxidant, and smooth muscle relaxant properties. A substantial decrease in the need for oxygen or intensive treatment assistance is reported in elderly COVID-19 patients upon the intake of vitamin D, magnesium, and vitamin B12 in combination 209. Iran’s clinical trial registry 210 confirmed that magnesium sulphate inhalation is effective in improving respiratory symptoms such as shortness of breath, cough and oxygen saturation in COVID-19 patients.

Magnesium deficient animal model exhibits inflammation as the first noticeable change with elevated levels of pro-inflammatory mediators like TNFα with declined anti-inflammatory cytokine levels 211, 201. The activation of immune cells like monocyte, macrophages, and polymorphonuclear cells are involved in the release of inflammatory mediators like cytokine, free radical and eicosanoids 201. Administration of magnesium reduces leukocyte activation and oxidative damage to peripheral blood lymphocyte DNA in athletes and sedentary young men 19. Thus, magnesium is an important factor for optimum immune cell functioning by regulating the proliferation and function of lymphocytes 198. In vitro studies also prove the role of magnesium in reducing leukocyte activation through its calcium antagonistic action 200. Magnesium deficiency results in the stress condition that activate the sympathetic system and hypothalamic-pituitary axis causes fat accumulation and release of neuropeptides; results in the immune response followed by inflammatory cascades 212.

Magnesium can inhibit cytokine storm and hyper oxidative stress in COVID-19 patients through various mechanisms such as its antioxidant, immune-modulatory, and anti-inflammatory activities 213.

Table 19. Magnesium Rich Foods

FoodMilligrams
(mg) per
serving
Percent
Daily Value (DV)*
Almonds, dry roasted, 1 ounce8020
Spinach, boiled, ½ cup7820
Cashews, dry roasted, 1 ounce7419
Peanuts, oil roasted, ¼ cup6316
Cereal, shredded wheat, 2 large biscuits6115
Soymilk, plain or vanilla, 1 cup6115
Black beans, cooked, ½ cup6015
Edamame, shelled, cooked, ½ cup5013
Peanut butter, smooth, 2 tablespoons4912
Bread, whole wheat, 2 slices4612
Avocado, cubed, 1 cup4411
Potato, baked with skin, 3.5 ounces4311
Rice, brown, cooked, ½ cup4211
Yogurt, plain, low fat, 8 ounces4211
Breakfast cereals, fortified with 10% of the DV for magnesium4010
Oatmeal, instant, 1 packet369
Kidney beans, canned, ½ cup359
Banana, 1 medium328
Salmon, Atlantic, farmed, cooked, 3 ounces267
Milk, 1 cup24–276–7
Halibut, cooked, 3 ounces246
Raisins, ½ cup236
Chicken breast, roasted, 3 ounces226
Beef, ground, 90% lean, pan broiled, 3 ounces205
Broccoli, chopped and cooked, ½ cup123
Rice, white, cooked, ½ cup103
Apple, 1 medium92
Carrot, raw, 1 medium72

Footnote: *DV = Daily Value. DVs were developed by the U.S. Food and Drug Administration (FDA) to help consumers compare the nutrient contents of products within the context of a total diet. The DV for magnesium is 400 mg for adults and children aged 4 and older. However, the FDA does not require food labels to list magnesium content unless a food has been fortified with this nutrient. Foods providing 20% or more of the DV are considered to be high sources of a nutrient.

[Source 214 ] References
  1. About Primary Immunodeficiencies. https://primaryimmune.org/about-primary-immunodeficiencies
  2. Marcos A. Editorial: A Review of Micronutrients and the Immune System-Working in Harmony to Reduce the Risk of Infection. Nutrients. 2021 Nov 22;13(11):4180. doi: 10.3390/nu13114180
  3. Gorji A, Khaleghi Ghadiri M. Potential roles of micronutrient deficiency and immune system dysfunction in the coronavirus disease 2019 (COVID-19) pandemic. Nutrition. 2021 Feb;82:111047. doi: 10.1016/j.nut.2020.111047
  4. James PT, Ali Z, Armitage AE, Bonell A, Cerami C, Drakesmith H, Jobe M, Jones KS, Liew Z, Moore SE, Morales-Berstein F, Nabwera HM, Nadjm B, Pasricha SR, Scheelbeek P, Silver MJ, Teh MR, Prentice AM. The Role of Nutrition in COVID-19 Susceptibility and Severity of Disease: A Systematic Review. J Nutr. 2021 Jul 1;151(7):1854-1878. doi: 10.1093/jn/nxab059
  5. Calder PC. Nutrition and immunity: lessons for COVID-19. Eur J Clin Nutr. 2021 Sep;75(9):1309-1318. doi: 10.1038/s41430-021-00949-8
  6. Calder PC, Carr AC, Gombart AF, Eggersdorfer M. Optimal Nutritional Status for a Well-Functioning Immune System Is an Important Factor to Protect against Viral Infections. Nutrients. 2020 Apr 23;12(4):1181. doi: 10.3390/nu12041181
  7. Gombart AF, Pierre A, Maggini S. A Review of Micronutrients and the Immune System-Working in Harmony to Reduce the Risk of Infection. Nutrients. 2020 Jan 16;12(1):236. doi: 10.3390/nu12010236
  8. Immunity In Brief. https://lpi.oregonstate.edu/mic/health-disease/immunity-in-brief
  9. Rice HB, Bernasconi A, Maki KC, Harris WS, von Schacky C, Calder PC. Conducting omega-3 clinical trials with cardiovascular outcomes: Proceedings of a workshop held at ISSFAL 2014. Prostaglandins Leukot Essent Fatty Acids. 2016 Apr;107:30-42. https://doi.org/10.1016/j.plefa.2016.01.003
  10. Skulas-Ray AC, Wilson PWF, Harris WS, Brinton EA, Kris-Etherton PM, Richter CK, Jacobson TA, Engler MB, Miller M, Robinson JG, Blum CB, Rodriguez-Leyva D, de Ferranti SD, Welty FK; American Heart Association Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Lifestyle and Cardiometabolic Health; Council on Cardiovascular Disease in the Young; Council on Cardiovascular and Stroke Nursing; and Council on Clinical Cardiology. Omega-3 Fatty Acids for the Management of Hypertriglyceridemia: A Science Advisory From the American Heart Association. Circulation. 2019 Sep 17;140(12):e673-e691. doi: 10.1161/CIR.0000000000000709
  11. Siscovick DS, Barringer TA, Fretts AM, Wu JH, Lichtenstein AH, Costello RB, Kris-Etherton PM, Jacobson TA, Engler MB, Alger HM, Appel LJ, Mozaffarian D; American Heart Association Nutrition Committee of the Council on Lifestyle and Cardiometabolic Health; Council on Epidemiology and Prevention; Council on Cardiovascular Disease in the Young; Council on Cardiovascular and Stroke Nursing; and Council on Clinical Cardiology. Omega-3 Polyunsaturated Fatty Acid (Fish Oil) Supplementation and the Prevention of Clinical Cardiovascular Disease: A Science Advisory From the American Heart Association. Circulation. 2017 Apr 11;135(15):e867-e884. doi: 10.1161/CIR.0000000000000482
  12. Rimm EB, Appel LJ, Chiuve SE, Djoussé L, Engler MB, Kris-Etherton PM, Mozaffarian D, Siscovick DS, Lichtenstein AH; American Heart Association Nutrition Committee of the Council on Lifestyle and Cardiometabolic Health; Council on Epidemiology and Prevention; Council on Cardiovascular Disease in the Young; Council on Cardiovascular and Stroke Nursing; and Council on Clinical Cardiology. Seafood Long-Chain n-3 Polyunsaturated Fatty Acids and Cardiovascular Disease: A Science Advisory From the American Heart Association. Circulation. 2018 Jul 3;138(1):e35-e47. doi: 10.1161/CIR.0000000000000574
  13. Maggini S., Pierre A., Calder P.C. Immune function and micronutrient requirements change over the life course. Nutrients. 2018;10:1531. doi: 10.3390/nu10101531
  14. Justiz Vaillant AA, Sabir S, Jan A. Physiology, Immune Response. [Updated 2020 Sep 27]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK539801
  15. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA) Guidance on the scientific requirements for health claims related to the immune system, the gastrointestinal tract and defence against pathogenic microorganisms. Efsa J 2016;14:4369.
  16. Forrest H Nielsen, Henry C Lukaski . Update on the relationship between magnesium and exercise. Magnesium Research. 2006;19(3):180-189. doi:10.1684/mrh.2006.0060
  17. F. C. Mooren, S.W. Golf, and K. Völker . Effect of magnesium on granulocyte function and on the exercise induced inflammatory response . Magnesium Research. 2003;16(1):49-58.
  18. Maria José Laires, Cristina Monteiro . Exercise, magnesium and immune function. Magnesium Research. 2008;21(2):92-96. doi:10.1684/mrh.2008.0136
  19. Petrović J, Stanić D, Dmitrašinović G, Plećaš-Solarović B, Ignjatović S, Batinić B, Popović D, Pešić V. Magnesium Supplementation Diminishes Peripheral Blood Lymphocyte DNA Oxidative Damage in Athletes and Sedentary Young Man. Oxid Med Cell Longev. 2016;2016:2019643. doi: 10.1155/2016/2019643
  20. Bussière FI, Mazur A, Fauquert JL, Labbe A, Rayssiguier Y, Tridon A. High magnesium concentration in vitro decreases human leukocyte activation. Magnes Res. 2002 Mar;15(1-2):43-8.
  21. Vitamins and minerals that affect the immune system. https://www.hiv.va.gov/patient/daily/diet/vitamin-mineral-chart.asp
  22. Imdad A, Mayo-Wilson E, Herzer K, et al. . Vitamin A supplementation for preventing morbidity and mortality in children from six months to five years of age. Cochrane Database Syst Rev 2017;3:CD008524. 10.1002/14651858.CD008524.pub3
  23. Hu N, QB L, Zou SY. Effect of vitamin A as an adjuvant therapy for pneumonia in children: a meta analysis. Zhongguo Dang Dai Er. Ke. Za Zhi 2018;20:146–53.
  24. Hemilä H, Louhiala P. Vitamin C for preventing and treating pneumonia. Cochrane Database Syst Rev 2013:CD005532. 10.1002/14651858.CD005532.pub3
  25. Hemilä H, Chalker E. Vitamin C for preventing and treating the common cold. Cochrane Database Syst Rev 2013:CD000980. 10.1002/14651858.CD000980.pub4
  26. Bergman P, Lindh Åsa U., Björkhem-Bergman L, et al. . Vitamin D and respiratory tract infections: a systematic review and meta-analysis of randomized controlled trials. PLoS One 2013;8:e65835 10.1371/journal.pone.0065835
  27. Martineau AR, Jolliffe DA, Hooper RL, et al. . Vitamin D supplementation to prevent acute respiratory tract infections: systematic review and meta-analysis of individual participant data. BMJ 2017;356:i6583 10.1136/bmj.i6583
  28. Pham H, Rahman A, Majidi A, et al. . Acute respiratory tract infection and 25-hydroxyvitamin D concentration: a systematic review and meta-analysis. Int J Environ Res Public Health 2019;16:3020 10.3390/ijerph16173020
  29. Zhou YF, Luo BA, Qin LL. The association between vitamin D deficiency and community-acquired pneumonia: a meta-analysis of observational studies. Medicine 2019;98:17252.
  30. Mao S, Zhang A, Huang S. Meta-Analysis of Zn, Cu and Fe in the hair of Chinese children with recurrent respiratory tract infection. Scand J Clin Lab Invest 2014;74:561–7. 10.3109/00365513.2014.921323
  31. Hemilä H. Zinc lozenges and the common cold: a meta-analysis comparing zinc acetate and zinc gluconate, and the role of zinc dosage. JRSM Open 2017;8:205427041769429. 10.1177/2054270417694291
  32. Science M, Johnstone J, Roth DE, et al. . Zinc for the treatment of the common cold: a systematic review and meta-analysis of randomized controlled trials. Can Med Assoc J 2012;184:E551–61. 10.1503/cmaj.111990
  33. Lassi ZS, Moin A, Bhutta ZA. Zinc supplementation for the prevention of pneumonia in children aged 2 months to 59 months. Cochrane Database Syst Rev 2016;12:CD005978. 10.1002/14651858.CD005978.pub3
  34. Wang L, Song Y. Efficacy of zinc given as an adjunct to the treatment of severe pneumonia: a meta-analysis of randomized, double-blind and placebo-controlled trials. Clin Respir J 2018;12:857–64. 10.1111/crj.12646
  35. Johnson EJ, Russell RM. Beta-Carotene. In: Coates PM, Betz JM, Blackman MR, et al., eds. Encyclopedia of Dietary Supplements. 2nd ed. London and New York: Informa Healthcare; 2010:115-20.
  36. Ross CA. Vitamin A. In: Coates PM, Betz JM, Blackman MR, et al., eds. Encyclopedia of Dietary Supplements. 2nd ed. London and New York: Informa Healthcare; 2010:778-91.
  37. Ross A. Vitamin A and Carotenoids. In: Shils M, Shike M, Ross A, Caballero B, Cousins R, eds. Modern Nutrition in Health and Disease. 10th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2006:351-75.
  38. Institute of Medicine, US Panel on Micronutrients. Dietary reference intakes for vitamin A, boron, chromium, copper, iodine, iron, manganese, molybdenum, nickel, silicon, vanadium, and zinc. National Academies Press. Washington, DC, 2001. PMID: 25057538 www.ncbi.nlm.nih.gov/pubmed/25057538.
  39. National Institute of Health. Vitamin A. https://ods.od.nih.gov/factsheets/VitaminA-HealthProfessional
  40. Oliveira LdeM, Teixeira FME, Sato MN. Impact of retinoic acid on immune cells and inflammatory diseases. Mediators Inflamm 2018;2018:1–17. 10.1155/2018/3067126
  41. Ross AC. Vitamin A deficiency and retinoid repletion regulate the antibody response to bacterial antigens and the maintenance of natural killer cells. Clin Immunol Immunopathol 1996;80:S63–72. 10.1006/clin.1996.0143
  42. Semba RD, Scott AL, et al. , Muhilal . Depressed immune response to tetanus in children with vitamin A deficiency. J Nutr 1992;122:101–7. 10.1093/jn/122.1.101
  43. U.S. Department of Agriculture, Agricultural Research Service. USDA National Nutrient Database for Standard Reference, Release 24. Nutrient Data Laboratory Home Page, 2011. https://www.ars.usda.gov/northeast-area/beltsville-md/beltsville-human-nutrition-research-center/nutrient-data-laboratory/
  44. Yoshii K, Hosomi K, Sawane K, et al. . Metabolism of dietary and microbial vitamin B family in the regulation of host immunity. Front. Nutr. 2019;6:48 10.3389/fnut.2019.00048
  45. Tamura J, Kubota K, Murakami H, et al. . Immunomodulation by vitamin B12: augmentation of CD8+ T lymphocytes and natural killer (NK) cell activity in vitamin B12-deficient patients by methyl-B12 treatment. Clin Exp Immunol 1999;116:28–32. 10.1046/j.1365-2249.1999.00870.x
  46. Meydani SN, Ribaya-Mercado JD, Russell RM, et al. . Vitamin B − 6 deficiency impairs interleukin 2 production and lymphocyte proliferation in elderly adults. Am J Clin Nutr 1991;53:1275–80. 10.1093/ajcn/53.5.1275
  47. Said HM. Thiamin. In: Coates PM, Betz JM, Blackman MR, et al., eds. Encyclopedia of Dietary Supplements. 2nd ed. London and New York: Informa Healthcare; 2010:748-53.
  48. U.S. Department of Agriculture, Agricultural Research Service. USDA National Nutrient Database for Standard Reference, Release 27. Nutrient Data Laboratory home page, 2014. https://ndb.nal.usda.gov/ndb/
  49. Said HM, Ross AC. Riboflavin. In: Ross AC, Caballero B, Cousins RJ, Tucker KL, Ziegler TR, eds. Modern Nutrition in Health and Disease. 11th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2014:325-30.
  50. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline 1998. https://www.nap.edu/catalog/6015/dietary-reference-intakes-for-thiamin-riboflavin-niacin-vitamin-b6-folate-vitamin-b12-pantothenic-acid-biotin-and-choline
  51. McCormick DB. Riboflavin. In: Erdman JW, Macdonald IA, Zeisel SH, eds. Present Knowledge in Nutrition. 10th ed. Washington, DC: Wiley-Blackwell; 2012:280-92.
  52. Rivlin RS. Riboflavin. In: Coates PM, Betz JM, Blackman MR, et al., eds. Encyclopedia of Dietary Supplements. 2nd ed. London and New York: Informa Healthcare; 2010:691-9.
  53. Gaylord AM, Warthesen JJ, Smith DE. Influence of milk fat, milk solids, and light intensity on the light stability of vitamin A and riboflavin in lowfat milk. J Dairy Sci 1986;69:2779-84. https://www.ncbi.nlm.nih.gov/pubmed/3805455?dopt=Abstract
  54. Mackey A, Davis S, Gregory J. Vitamin B6. In: Shils M, Shike M, Ross A, Caballero B, Cousins R, eds. Modern Nutrition in Health and Disease. 10th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2005.
  55. McCormick D. Vitamin B6. In: Bowman B, Russell R, eds. Present Knowledge in Nutrition. 9th ed. Washington, DC: International Life Sciences Institute; 2006.
  56. Herbert V. Vitamin B12 in Present Knowledge in Nutrition. 17th ed. Washington, DC: International Life Sciences Institute Press, 1996.
  57. Herbert V, Das K. Vitamin B12 in Modern Nutrition in Health and Disease. 8th ed. Baltimore, MD: Williams & Wilkins, 1994.
  58. Combs G. Vitamin B12 in The Vitamins. New York: Academic Press, Inc., 1992.
  59. Zittoun J, Zittoun R. Modern clinical testing strategies in cobalamin and folate deficiency. Sem Hematol 1999;36:35-46. https://www.ncbi.nlm.nih.gov/pubmed/9930567?dopt=Abstract
  60. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes: Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. Washington, DC: National Academy Press, 1998.
  61. Klee GG. Cobalamin and folate evaluation: measurement of methylmalonic acid and homocysteine vs vitamin B(12) and folate. Clin Chem 2000;46:1277-83. https://www.ncbi.nlm.nih.gov/pubmed/10926922?dopt=Abstract
  62. Carmel R. How I treat cobalamin (vitamin B12) deficiency. Blood.2008;112:2214-21. https://www.ncbi.nlm.nih.gov/pubmed/18606874?dopt=Abstract
  63. U.S. National Library of Medicine, MedlinePlus – Folic Acid – https://medlineplus.gov/folicacid.html
  64. Office on Women’s Health, U.S. Department of Health and Human Services – Folic acid – https://www.womenshealth.gov/a-z-topics/folic-acid
  65. Centers for Disease Control and Prevention – Facts About Folic Acid – https://www.cdc.gov/ncbddd/folicacid/about.html
  66. Naidu KA: Vitamin C in human health and disease is still a mystery? An overview. Nutr J 2: 7, 2003. https://www.ncbi.nlm.nih.gov/pubmed/14498993?dopt=Abstract
  67. Li Y, Schellhorn HE. New developments and novel therapeutic perspectives for vitamin C. J Nutr 2007;137:2171-84. https://www.ncbi.nlm.nih.gov/pubmed/17884994?dopt=Abstract
  68. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids Washington, DC: National Academy Press; 2000.
  69. Jacob RA, Sotoudeh G. Vitamin C function and status in chronic disease. Nutr Clin Care 2002;5:66-74. https://www.ncbi.nlm.nih.gov/pubmed/12134712
  70. Merck Sharp & Dohme Corp., Merck Manual. Vitamin C (Ascorbic Acid). https://www.merckmanuals.com/professional/nutritional-disorders/vitamin-deficiency,-dependency,-and-toxicity/vitamin-c
  71. Holford P, Carr AC, Jovic TH, Ali SR, Whitaker IS, Marik PE, Smith AD. Vitamin C-An Adjunctive Therapy for Respiratory Infection, Sepsis and COVID-19. Nutrients. 2020 Dec 7;12(12):3760. doi: 10.3390/nu12123760
  72. Carr AC, Maggini S. Vitamin C and Immune Function. Nutrients. 2017 Nov 3;9(11):1211. doi: 10.3390/nu9111211
  73. Hemilä H, Chalker E. Vitamin C for preventing and treating the common cold. Cochrane Database Syst Rev. 2013 Jan 31;2013(1):CD000980. doi: 10.1002/14651858.CD000980.pub4
  74. Johnston CS. Vitamin C. In: Marriott BP, Birt DF, Stallings VA, Yates AA, eds. Present Knowledge in Nutrition 11th ed. Cambridge, MA: Elsevier; 2020:155-69.
  75. Hemilä H. Vitamin C and Infections. Nutrients. 2017 Mar 29;9(4):339. doi: 10.3390/nu9040339
  76. Schleicher RL, Carroll MD, Ford ES, Lacher DA. Serum vitamin C and the prevalence of vitamin C deficiency in the United States: 2003-2004 National Health and Nutrition Examination Survey (NHANES). Am J Clin Nutr. 2009 Nov;90(5):1252-63. doi: 10.3945/ajcn.2008.27016
  77. National Cancer Institute. High-Dose Vitamin C–Patient Version. https://www.cancer.gov/about-cancer/treatment/cam/patient/vitamin-c-pdq#link/_5
  78. Hemilä H. Vitamin C and infections. Nutrients 2017;9:339 10.3390/nu9040339
  79. Jacob RA, Kelley DS, Pianalto FS, et al. . Immunocompetence and oxidant defense during ascorbate depletion of healthy men. Am J Clin Nutr 1991;54:1302S–9. 10.1093/ajcn/54.6.1302s
  80. Carr AC, Frei B. Toward a new recommended dietary allowance for vitamin C based on antioxidant and health effects in humans. Am J Clin Nutr 1999;69:1086-107. https://www.ncbi.nlm.nih.gov/pubmed/10357726?dopt=Abstract
  81. Proc Natl Acad Sci U S A. 1989 Aug;86(16):6377-81. Ascorbate is an outstanding antioxidant in human blood plasma. https://www.ncbi.nlm.nih.gov/pubmed/2762330%20?dopt=Abstract
  82. Jacob RA, Sotoudeh G. Vitamin C function and status in chronic disease. Nutr Clin Care 2002;5:66-74. https://www.ncbi.nlm.nih.gov/pubmed/12134712?dopt=Abstract
  83. Gershoff SN. Vitamin C (ascorbic acid): new roles, new requirements? Nutr Rev 1993;51:313-26. https://www.ncbi.nlm.nih.gov/pubmed/8108031?dopt=Abstract
  84. Weinstein M, Babyn P, Zlotkin S. An orange a day keeps the doctor away: scurvy in the year 2000. Pediatrics 2001;108:E55. https://www.ncbi.nlm.nih.gov/pubmed/11533373?dopt=Abstract
  85. Wang AH, Still C. Old world meets modern: a case report of scurvy. Nutr Clin Pract 2007;22:445-8. https://www.ncbi.nlm.nih.gov/pubmed/17644699?dopt=Abstract
  86. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. Washington, DC: National Academy Press, 2000. https://www.nap.edu/catalog/9810/dietary-reference-intakes-for-vitamin-c-vitamin-e-selenium-and-carotenoids
  87. Stephen R, Utecht T. Scurvy identified in the emergency department: a case report. J Emerg Med 2001;21:235-7. https://www.ncbi.nlm.nih.gov/pubmed/11604276
  88. National Institute of Health. Vitamin D. https://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional/
  89. Institute of Medicine, Food and Nutrition Board. Dietary Reference Intakes for Calcium and Vitamin D. Washington, DC: National Academy Press, 2010
  90. Cranney C, Horsely T, O’Donnell S, Weiler H, Ooi D, Atkinson S, et al. Effectiveness and safety of vitamin D. Evidence Report/Technology Assessment No. 158 prepared by the University of Ottawa Evidence-based Practice Center under Contract No. 290-02.0021. AHRQ Publication No. 07-E013. Rockville, MD: Agency for Healthcare Research and Quality, 2007. https://www.ncbi.nlm.nih.gov/pubmed/18088161?dopt=Abstract
  91. Holick MF. Vitamin D. In: Shils ME, Shike M, Ross AC, Caballero B, Cousins RJ, eds. Modern Nutrition in Health and Disease, 10th ed. Philadelphia: Lippincott Williams & Wilkins, 2006.
  92. Norman AW, Henry HH. Vitamin D. In: Bowman BA, Russell RM, eds. Present Knowledge in Nutrition, 9th ed. Washington DC: ILSI Press, 2006.
  93. Medrano M, Carrillo-Cruz E, Montero I, et al. . Vitamin D: effect on haematopoiesis and immune system and clinical applications. Int J Mol Sci 2018;19:2663 10.3390/ijms19092663
  94. Gombart AF. The vitamin D-antimicrobial peptide pathway and its role in protection against infection. Future Microbiol 2009;4:1151–65. 10.2217/fmb.09.87
  95. Lee M-D, Lin C-H, Lei W-T, et al. . Does vitamin D deficiency affect the immunogenic responses to influenza vaccination? A systematic review and meta-analysis. Nutrients 2018;10:409 10.3390/nu10040409
  96. Berry DJ, Hesketh K, Power C, et al. . Vitamin D status has a linear association with seasonal infections and lung function in British adults. Br J Nutr 2011;106:1433–40. 10.1017/S0007114511001991
  97. Ginde AA, Mansbach JM, Camargo CA. Association between serum 25-hydroxyvitamin D level and upper respiratory tract infection in the third National Health and Nutrition Examination Survey. Arch Intern Med 2009;169:384–90. 10.1001/archinternmed.2008.560
  98. Sabetta JR, DePetrillo P, Cipriani RJ, et al. . Serum 25-hydroxyvitamin D and the incidence of acute viral respiratory tract infections in healthy adults. PLoS One 2010;5:e11088 10.1371/journal.pone.0011088
  99. Urashima M, Segawa T, Okazaki M, et al. . Randomized trial of vitamin D supplementation to prevent seasonal influenza A in schoolchildren. Am J Clin Nutr 2010;91:1255–60. 10.3945/ajcn.2009.29094
  100. Traber MG. Vitamin E. In: Shils ME, Shike M, Ross AC, Caballero B, Cousins R, eds. Modern Nutrition in Health and Disease. 10th ed. Baltimore, MD: Lippincott Williams & Wilkins, 2006;396-411.
  101. Merck Sharp & Dohme Corp., Merck Manual. Vitamin E (Tocopherol). https://www.merckmanuals.com/professional/nutritional-disorders/vitamin-deficiency,-dependency,-and-toxicity/vitamin-e
  102. Verhagen H, Buijsse B, Jansen E, Bueno-de-Mesquita B. The state of antioxidant affairs. Nutr Today 2006;41:244-50.
  103. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes: Vitamin C, Vitamin E, Selenium, and Carotenoids. Washington, DC: National Academy Press, 2000. https://www.nap.edu/catalog/9810/dietary-reference-intakes-for-vitamin-c-vitamin-e-selenium-and-carotenoids
  104. Lee G, Han S. The role of vitamin E in immunity. Nutrients 2018;10:614 10.3390/nu10111614
  105. Chavance M, Herbeth B, Fournier C, et al. . Vitamin status, immunity and infections in an elderly population. Eur J Clin Nutr 1989;43:827–35.
  106. De la Fuente M, Hernanz A, Guayerbas N, et al. . Vitamin E ingestion improves several immune functions in elderly men and women. Free Radic Res 2008;42:272–80. 10.1080/10715760801898838
  107. Meydani SN, et al. Vitamin E supplementation and in vivo immune response in healthy elderly subjects. JAMA 1997;277:1380–6. 10.1001/jama.1997.03540410058031
  108. Meydani SN, Barklund MP, Liu S, et al. . Vitamin E supplementation enhances cell-mediated immunity in healthy elderly subjects. Am J Clin Nutr 1990;52:557–63. 10.1093/ajcn/52.3.557
  109. Hemilä H. Vitamin E administration may decrease the incidence of pneumonia in elderly males. Clin Interv Aging 2016;11:1379–85. 10.2147/CIA.S114515
  110. Meydani SN, Leka LS, Fine BC, et al. . Vitamin E and respiratory tract infections in elderly nursing home residents: a randomized controlled trial. JAMA 2004;292:828–36. 10.1001/jama.292.7.828
  111. Graat JM, Schouten EG, Kok FJ. Effect of daily vitamin E and multivitamin-mineral supplementation on acute respiratory tract infections in elderly persons: a randomized controlled trial. JAMA 2002;288:715–21. 10.1001/jama.288.6.715
  112. U.S. National Library of Medicine. Medline Plus. Iron. https://medlineplus.gov/iron.html#cat_51
  113. Centers for Disease Control and Prevention. Recommendations to Prevent and Control Iron Deficiency in the United States. https://www.cdc.gov/mmwr/preview/mmwrhtml/00051880.htm
  114. U.S. National Library of Medicine. Iron. https://livertox.nlm.nih.gov/Iron.htm
  115. Nairz M, Dichtl S, Schroll A, et al. . Iron and innate antimicrobial immunity-Depriving the pathogen, defending the host. J Trace Elem Med Biol 2018;48:118–33. 10.1016/j.jtemb.2018.03.007
  116. Ahluwalia N, Sun J, Krause D, et al. . Immune function is impaired in iron-deficient, homebound, older women. Am J Clin Nutr 2004;79:516–21. 10.1093/ajcn/79.3.516
  117. Nairz M, Theurl I, Swirski FK, et al. . “Pumping iron”—how macrophages handle iron at the systemic, microenvironmental, and cellular levels. Pflugers Arch – Eur J Physiol 2017;469:397–418. 10.1007/s00424-017-1944-8
  118. Ganz T. Iron and infection. Int J Hematol 2018;107:7–15. 10.1007/s12185-017-2366-2
  119. Malan L, Baumgartner J, Calder PC, et al. . n–3 long-chain PUFAs reduce respiratory morbidity caused by iron supplementation in iron-deficient South African schoolchildren: a randomized, double-blind, placebo-controlled intervention. Am J Clin Nutr 2015;101:668–79. 10.3945/ajcn.113.081208
  120. Murray-Kolbe LE, Beard J. Iron. In: Coates PM, Betz JM, Blackman MR, et al., eds. Encyclopedia of Dietary Supplements. 2nd ed. London and New York: Informa Healthcare; 2010:432-8.
  121. Aggett PJ. Iron. In: Erdman JW, Macdonald IA, Zeisel SH, eds. Present Knowledge in Nutrition. 10th ed. Washington, DC: Wiley-Blackwell; 2012:506-20.
  122. Hurrell R, Egli I. Iron bioavailability and dietary reference values. Am J Clin Nutr 2010;91:1461S-7S. http://ajcn.nutrition.org/content/91/5/1461S.long
  123. Rutzke CJ, Glahn RP, Rutzke MA, Welch RM, Langhans RW, Albright LD, et al. Bioavailability of iron from spinach using an in vitro/human Caco-2 cell bioassay model. Habitation 2004;10:7-14. https://www.ncbi.nlm.nih.gov/pubmed/15880905
  124. Collins JF. Copper. In: Ross AC, Caballero B, Cousins RJ, Tucker KL, Ziegler TR, eds. Modern Nutrition in Health and Disease. 11th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2014:206-16.
  125. Allen KG, Klevay LM. Copper: an antioxidant nutrient for cardiovascular health. Curr Opin Lipidol. 1994 Feb;5(1):22-8. doi: 10.1097/00041433-199402000-00005
  126. Hellman NE, Gitlin JD. Ceruloplasmin metabolism and function. Annu Rev Nutr. 2002;22:439-58. doi: 10.1146/annurev.nutr.22.012502.114457
  127. Li C, Li Y, Ding C. The role of copper homeostasis at the host-pathogen axis: from bacteria to fungi. Int J Mol Sci 2019;20:175 10.3390/ijms20010175
  128. Hopkins RG, Failla ML. Copper deficiency reduces interleukin-2 (IL-2) production and IL-2 mRNA in human T-lymphocytes. J Nutr 1997;127:257–62. 10.1093/jn/127.2.257
  129. Vyas D, Chandra RK. Thymic factor activity, lymphocyte stimulation response and antibody producing cells in copper deficiency. Nutr Res 1983;3:343–9. 10.1016/S0271-5317(83)80084-0
  130. Klevay LM. Is the Western diet adequate in copper? J Trace Elem Med Biol. 2011 Dec;25(4):204-12. doi: 10.1016/j.jtemb.2011.08.146
  131. Institute of Medicine, Food and Nutrition Board. Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc. Washington, DC: National Academies Press; 2001.
  132. Prohaska JR. Copper. In: Erdman JW, Macdonald IA, Zeisel SH, eds. Present Knowledge in Nutrition. 10th ed. Washington, DC: Wiley-Blackwell; 2012:540-53.
  133. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes: Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. Washington, DC: National Academy Press; 1998.
  134. U.S. Department of Agriculture, Agricultural Research Service. What We Eat in America, 2013-2014. https://www.ars.usda.gov/northeast-area/beltsville-md-bhnrc/beltsville-human-nutrition-research-center/food-surveys-research-group/docs/wweia-data-tables
  135. Prohaska JR. Impact of copper deficiency in humans. Ann N Y Acad Sci. 2014 May;1314:1-5. doi: 10.1111/nyas.12354
  136. National Institutes of Health. Dietary Supplement Label Database. 2021. https://dsld.od.nih.gov/dsld
  137. Rosado JL. Zinc and copper: proposed fortification levels and recommended zinc compounds. J Nutr. 2003 Sep;133(9):2985S-9S. doi: 10.1093/jn/133.9.2985S
  138. Sunde RA. Selenium. In: Ross AC, Caballero B, Cousins RJ, Tucker KL, Ziegler TR, eds. Modern Nutrition in Health and Disease. 11th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2012:225-37
  139. Sunde RA. Selenium. In: Bowman B, Russell R, eds. Present Knowledge in Nutrition. 9th ed. Washington, DC: International Life Sciences Institute; 2006:480-97
  140. Terry EN, Diamond AM. Selenium. In: Erdman JW, Macdonald IA, Zeisel SH, eds. Present Knowledge in Nutrition. 10th ed. Washington, DC: Wiley-Blackwell; 2012:568-87
  141. Environ Health Prev Med. 2008 Mar; 13(2): 102–108. Selenium: its role as antioxidant in human health. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2698273/
  142. Br J Nutr. 2008 Aug;100(2):254-68. doi: 10.1017/S0007114508939830. Epub 2008 Mar 18. Food-chain selenium and human health: emphasis on intake. https://www.ncbi.nlm.nih.gov/pubmed/18346308?dopt=Abstract
  143. Lancet. 2012 Mar 31;379(9822):1256-68. doi: 10.1016/S0140-6736(11)61452-9. Epub 2012 Feb 29. Selenium and human health. https://www.ncbi.nlm.nih.gov/pubmed/22381456?dopt=Abstract
  144. Guillin OM, Vindry C, Ohlmann T, et al. . Selenium, selenoproteins and viral infection. Nutrients 2019;11:2101 10.3390/nu11092101
  145. Beck M, Handy J, Levander O. Host nutritional status: the neglected virulence factor. Trends Microbiol 2004;12:417–23. 10.1016/j.tim.2004.07.007
  146. Hawkes WC, Kelley DS, Taylor PC. The effects of dietary selenium on the immune system in healthy men. Biol Trace Elem Res 2001;81:189–213. 10.1385/BTER:81:3:189
  147. Roy M, Kiremidjian-Schumacher L, Wishe HI, et al. . Supplementation with selenium restores age-related decline in immune cell function. Exp Biol Med 1995;209:369–75. 10.3181/00379727-209-43909
  148. Broome CS, McArdle F, Kyle JAM, et al. . An increase in selenium intake improves immune function and poliovirus handling in adults with marginal selenium status. Am J Clin Nutr 2004;80:154–62. 10.1093/ajcn/80.1.154
  149. https://ndb.nal.usda.gov/ndb/
  150. Sandstead HH. Understanding zinc: recent observations and interpretations. J Lab Clin Med 1994;124:322-7. https://www.ncbi.nlm.nih.gov/pubmed/8083574?dopt=Abstract
  151. Institute of Medicine, Food and Nutrition Board. Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc (2001). Washington, DC: National Academy Press, 2001. https://www.nap.edu/catalog/10026/dietary-reference-intakes-for-vitamin-a-vitamin-k-arsenic-boron-chromium-copper-iodine-iron-manganese-molybdenum-nickel-silicon-vanadium-and-zinc
  152. Solomons NW. Mild human zinc deficiency produces an imbalance between cell-mediated and humoral immunity. Nutr Rev 1998;56:27-8. https://www.ncbi.nlm.nih.gov/pubmed/9481116?dopt=Abstract
  153. Prasad AS. Zinc: an overview. Nutrition 1995;11:93-9. https://www.ncbi.nlm.nih.gov/pubmed/7749260?dopt=Abstract
  154. Heyneman CA. Zinc deficiency and taste disorders. Ann Pharmacother 1996;30:186-7. https://www.ncbi.nlm.nih.gov/pubmed/8835055?dopt=Abstract
  155. Simmer K, Thompson RP. Zinc in the fetus and newborn. Acta Paediatr Scand Suppl 1985;319:158-63. https://www.ncbi.nlm.nih.gov/pubmed/3868917?dopt=Abstract
  156. Fabris N, Mocchegiani E. Zinc, human diseases and aging. Aging (Milano) 1995;7:77-93. https://www.ncbi.nlm.nih.gov/pubmed/7548268?dopt=Abstract
  157. Maret W, Sandstead HH. Zinc requirements and the risks and benefits of zinc supplementation. J Trace Elem Med Biol 2006;20:3-18. https://www.ncbi.nlm.nih.gov/pubmed/16632171?dopt=Abstract
  158. Prasad AS, Beck FW, Grabowski SM, Kaplan J, Mathog RH. Zinc deficiency: changes in cytokine production and T-cell subpopulations in patients with head and neck cancer and in noncancer subjects. Proc Assoc Am Physicians 1997;109:68-77. https://www.ncbi.nlm.nih.gov/pubmed/9010918?dopt=Abstract
  159. Rink L, Gabriel P. Zinc and the immune system. Proc Nutr Soc 2000;59:541-52. https://www.ncbi.nlm.nih.gov/pubmed/11115789?dopt=Abstract
  160. Maywald M, Wessels I, Rink L. Zinc signals and immunity. Int J Mol Sci 2017;18:2222 10.3390/ijms18102222
  161. Read SA, Obeid S, Ahlenstiel C, et al. . The role of zinc in antiviral immunity. Adv Nutr 2019;10:696–710. 10.1093/advances/nmz013
  162. Kaushik N, Subramani C, Anang S, et al. . Zinc salts block hepatitis E virus replication by inhibiting the activity of viral RNA-dependent RNA polymerase. J. Virol 2017;91i:e00754–17
  163. Uchide N, Ohyama K, Bessho T, et al. . Effect of antioxidants on apoptosis induced by influenza virus infection: inhibition of viral gene replication and transcription with pyrrolidine dithiocarbamate. Antiviral Res 2002;56:207–17. 10.1016/S0166-3542(02)00109-2
  164. te Velthuis AJW, van den Worm SHE, Sims AC, et al. . Zn2+ inhibits coronavirus and arterivirus RNA polymerase activity in vitro and zinc ionophores block the replication of these viruses in cell culture. PLoS Pathog 2010;6:e1001176 10.1371/journal.ppat.1001176
  165. Subramanian Vignesh K, Deepe Jr. G. Metallothioneins: emerging modulators in immunity and infection. Int J Mol Sci 2017;18:2197 10.3390/ijms18102197
  166. Hasan R, Rink L, Haase H. Zinc signals in neutrophil granulocytes are required for the formation of neutrophil extracellular traps. Innate Immun 2013;19:253–64. 10.1177/1753425912458815
  167. Hasan R, Rink L, Haase H. Chelation of free Zn²⁺ impairs chemotaxis, phagocytosis, oxidative burst, degranulation, and cytokine production by neutrophil granulocytes. Biol Trace Elem Res 2016;171:79–88. 10.1007/s12011-015-0515-0
  168. Kahmann L, Uciechowski P, Warmuth S, et al. . Zinc supplementation in the elderly reduces spontaneous inflammatory cytokine release and restores T cell functions. Rejuvenation Res 2008;11:227–37. 10.1089/rej.2007.0613
  169. Tapazoglou E, Prasad AS, Hill G, et al. . Decreased natural killer cell activity in patients with zinc deficiency with sickle cell disease. J Lab Clin Med 1985;105:19–22.
  170. Sandström B, et al. Acrodermatitis enteropathica, zinc metabolism, copper status, and immune function. Arch Pediatr Adolesc Med 1994;148:980–5. 10.1001/archpedi.1994.02170090094017
  171. Barnett JB, Dao MC, Hamer DH, et al. . Effect of zinc supplementation on serum zinc concentration and T cell proliferation in nursing home elderly: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr 2016;103:942–51. 10.3945/ajcn.115.115188
  172. Gammoh NZ, Rink L. Zinc in infection and inflammation. Nutrients 2017;9:624. 10.3390/nu9060624
  173. National Institutes of Health. Magnesium. https://ods.od.nih.gov/factsheets/Magnesium-HealthProfessional/
  174. Institute of Medicine (IOM). Food and Nutrition Board. Dietary Reference Intakes: Calcium, Phosphorus, Magnesium, Vitamin D and Fluoride. Washington, DC: National Academy Press, 1997.
  175. Rude RK. Magnesium. In: Coates PM, Betz JM, Blackman MR, Cragg GM, Levine M, Moss J, White JD, eds. Encyclopedia of Dietary Supplements. 2nd ed. New York, NY: Informa Healthcare; 2010:527-37.
  176. Rude RK. Magnesium. In: Ross AC, Caballero B, Cousins RJ, Tucker KL, Ziegler TR, eds. Modern Nutrition in Health and Disease. 11th ed. Baltimore, Mass: Lippincott Williams & Wilkins; 2012:159-75.
  177. Pham PCT, Pham PAT, Pham SV, Pham PTT, Pham PMT, Pham PTT. Hypomagnesemia: a clinical perspective. Int J Nephrol Renovasc Dis. 2014 doi: 10.2147/IJNRD.S42054
  178. Bharadwaj J, Darbari A, Naithani M. Magnesium: the fifth electrolyte. J Med Nutr Nutraceuticals. 2014;3:186. doi: 10.4103/2278-019x.131955
  179. Ahmed F, Mohammed A. Magnesium: the forgotten electrolyte—a review on hypomagnesemia. Med Sci. 2019;7:56. doi: 10.3390/medsci7040056
  180. Fiorentini D, Cappadone C, Farruggia G, Prata C. Magnesium: Biochemistry, Nutrition, Detection, and Social Impact of Diseases Linked to Its Deficiency. Nutrients. 2021 Mar 30;13(4):1136. doi: 10.3390/nu13041136
  181. Volpe SL. Magnesium. In: Erdman JW, Macdonald IA, Zeisel SH, eds. Present Knowledge in Nutrition. 10th ed. Ames, Iowa; John Wiley & Sons, 2012:459-74.
  182. Mathew AA, Panonnummal R. ‘Magnesium’-the master cation-as a drug-possibilities and evidences. Biometals. 2021 Oct;34(5):955-986. doi: 10.1007/s10534-021-00328-7
  183. Al Alawi AM, Majoni SW, Falhammar H. Magnesium and human health: perspectives and research directions. Int J Endocrinol. 2018 doi: 10.1155/2018/9041694
  184. Elin RJ. Assessment of magnesium status for diagnosis and therapy. Magnes Res 2010;23:1-5. https://www.ncbi.nlm.nih.gov/pubmed/20736141?dopt=Abstract
  185. Gibson, RS. Principles of Nutritional Assessment, 2nd ed. New York, NY: Oxford University Press, 2005.
  186. Alexander RT, Hoenderop JG, Bindels RJ. Molecular determinants of magnesium homeostasis: insights from human disease. J Am Soc Nephrol. 2008;19:1451–1458. doi: 10.1681/ASN.2008010098
  187. Dolati S, Rikhtegar R, Mehdizadeh A, Yousefi M. The role of magnesium in pathophysiology and migraine treatment. Biol Trace Elem Res. 2019 doi: 10.1007/s12011-019-01931-z
  188. Gröber U, Schmidt J, Kisters K. Magnesium in prevention and therapy. Nutrients. 2015;7:8199–8226. doi: 10.3390/nu7095388
  189. DiNicolantonio JJ, O’Keefe JH, Wilson W. Subclinical magnesium deficiency: a principal driver of cardiovascular disease and a public health crisis. Open Hear. 2018 doi: 10.1136/openhrt-2017-000668
  190. Li X, Han X, Yang J, Bao J, Di X, Zhang G, Liu H. Magnesium sulfate provides neuroprotection in eclampsia-like seizure model by ameliorating neuroinflammation and brain edema. Mol Neurobiol. 2017;54:7938–7948. doi: 10.1007/s12035-016-0278-4
  191. Euser AG, Cipolla MJ. Magnesium sulfate for the treatment of eclampsia: a brief review. Stroke. 2009;40:1169–1175. doi: 10.1161/STROKEAHA.108.527788
  192. Witkowski M, Hubert J, Mazur A. Methods of assessment of magnesium status in humans: a systematic review. Magnesium Res 2011;24:163-80. https://www.ncbi.nlm.nih.gov/pubmed/22064327?dopt=Abstract
  193. Tam M, Gómez S, González-Gross M, Marcos A. Possible roles of magnesium on the immune system. Eur J Clin Nutr. 2003;57:1193–1197. doi: 10.1038/sj.ejcn.1601689
  194. Schlingmann KP, Waldegger S, Konrad M, Chubanov V, Gudermann T. TRPM6 and TRPM7-gatekeepers of human magnesium metabolism. Biochim Biophys Acta. 2007;1772:813–821. doi: 10.1016/j.bbadis.2007.03.009
  195. Ravell J, Chaigne-Delalande B, Lenardo M. X-linked immunodeficiency with magnesium defect, Epstein-Barr virus infection, and neoplasia disease: a combined immune deficiency with magnesium defect. Curr Opin Pediatr. 2014;26:713–719. doi: 10.1097/MOP.0000000000000156
  196. Li FY, Chaigne-Delalande B, Kanellopoulou C, Davis JC, Matthews HF, Douek DC, Cohen JI, Uzel G, Su HC, Lenardo MJ. Second messenger role for Mg2+ revealed by human T-cell immunodeficiency. Nature. 2011 Jul 27;475(7357):471-6. doi: 10.1038/nature10246
  197. Liang RY, Wu W, Huang J, Jiang SP, Lin Y. Magnesium affects the cytokine secretion of CD4+ T lymphocytes in acute asthma. J Asthma. 2012;49:1012–1015. doi: 10.3109/02770903.2012.739240
  198. Kubena KS. The role of magnesium in immunity. J Nutr Immunol. 1994;2(3):107–126. doi: 10.1300/J053v02n03_07
  199. Mazur A., Maier J.A., Rock E., Gueux E., Nowacki W., Rayssiguier Y. Magnesium and the inflammatory response: Potential physiopathological implications. Arch. Biochem. Biophys. 2007;458:48–56. doi: 10.1016/j.abb.2006.03.031
  200. Laires MJ, Monteiro C. Exercise, magnesium and immune function. Magnes Res. 2008;21(2):92–96. doi: 10.1684/mrh.2008.0136
  201. Galland L. Magnesium and immune function: an overview. Magnesium. 1988;7(5-6):290-9.
  202. Nielsen FH. Magnesium deficiency and increased inflammation: current perspectives. J Inflamm Res. 2018;11:25–34. doi: 10.2147/JIR.S136742
  203. Nielsen F.H. Magnesium deficiency and increased inflammation: Current perspectives. J. Inflamm. Res. 2018;11:25–34. doi: 10.2147/JIR.S136742
  204. Klein G.L. The Role of Calcium in Inflammation-Associated Bone Resorption. Biomolecules. 2018;8:69. doi: 10.3390/biom8030069
  205. Güzel A., Doğan E., Türkçü G., Kuyumcu M., Kaplan İ., Çelik F., Yıldırım Z.B. Dexmedetomidine and Magnesium Sulfate: A Good Combination Treatment for Acute Lung Injury? J. Investig. Surg. 2019;32:331–342. doi: 10.1080/08941939.2017.1422575
  206. Tang C.-F., Ding H., Jiao R.-Q., Wu X.-X., Kong L.-D. Possibility of magnesium supplementation for supportive treatment in patients with COVID-19. Eur. J. Pharmacol. 2020;886:173546. doi: 10.1016/j.ejphar.2020.173546
  207. Iotti S., Wolf F., Mazur A., Maier J.A. The COVID-19 pandemic: Is there a role for magnesium? Hypotheses and perspectives. Magnes. Res. 2020;33:21–27. doi: 10.1684/mrh.2020.0465
  208. Tang CF, Ding H, Jiao RQ, Xing Xin Wu, Kong LD. Possibility of magnesium supplementation for supportive treatment in patients with COVID-19. Eur J Pharmacol. 2020;886:173546. doi: 10.1016/j.ejphar.2020.173546
  209. Tan CW, Ho LP, Kalimuddin S, Cherng BPZ, Teh YE, Thien SY, Wong HM, Tern PJW, et al. A cohort study to evaluate the effect of combination vitamin D, magnesium and vitamin B12 (DMB) on progression to severe outcome in older COVID-19 patients. MedRxiv. 2020 doi: 10.1101/2020.06.01.20112334
  210. Pourdowlat G, Mousavinasab SR, Farzanegan B, Kashefizadeh A, Meybodi ZA, Jafarzadeh M, Baniasadi S. Evaluation of the efficacy and safety of inhaled magnesium sulphate in combination with standard treatment in patients with moderate or severe COVID-19: a structured summary of a study protocol for a randomised controlled trial. Trials. 2021;22:60. doi: 10.1186/s13063-021-05032-y
  211. Schmitz C, Anne-Laure P. Molecular, genetic, and nutritional aspects of major and trace minerals. In: Collins J, editor. Magnesium and the immune response. Amsterdam: Elsevier Inc; 2017. pp. 319–331.
  212. Rayssiguier Y, Libako P, Nowacki W, Rock E. Magnesium deficiency and metabolic syndrome: stress and inflammation may reflect calcium activation. Magnes Res. 2010;23(2):73–80. doi: 10.1684/mrh.2010.0208
  213. Moh’d Nour MBY, Alshawabkeh AD, Jadallah ARR, et al. Magnesium sulfate extended infusion as an adjunctive treatment for complicated Covid-19 infected critically ill patients. EAS J Anesthesiol Crit Care. 2020 doi: 10.36349/easjacc.2020.v02i03.17
  214. United States Department of Agriculture Agricultural Research Service. USDA Food Composition Databases. https://ndb.nal.usda.gov/ndb/
read more
BloodConditions & DiseasesImmune and Hematology System

Anemia of prematurity

anemia of prematurity

Anemia of prematurity

Anemia of prematurity means that a baby born early (prematurely) does not have enough red blood cells. Red blood cells carry oxygen to the body. Preterm infants with birth weight <1.0 kg (commonly designated as extremely low birth weight or ELBW, infants) have completed ≤29 weeks of gestation, and nearly all will need red blood cell (RBC) transfusions during the first weeks of life. Every week in the United States, approximately 10,000 infants are born prematurely (ie, <37 weeks of gestation), with 600 (6%) of these preterm infants being extremely low birth weight 1. Approximately 90% of extremely low birth weight neonates will receive at least one red blood cell transfusion 2.

All babies have some anemia (decrease in hemoglobin concentration) when they are born. In healthy term infants, the nadir hemoglobin value rarely falls below 10 g/dL at an age of 10 to 12 weeks 3. This is normal and is called “physiological anemia of infancy”. For the term infant, a physiologic and usually asymptomatic anemia is observed 8-12 weeks after birth. But in premature babies, the number of red blood cells may decrease faster and go lower than in full-term babies. This may happen because:

  • A premature baby may not make enough red blood cells.
  • A premature baby may need tests that require blood samples. It may be hard for the baby to produce enough red blood cells to make up for the blood that’s taken out and used in the tests.
  • A baby’s red blood cells don’t live as long as an older child’s red blood cells.

Anemia of prematurity is an exaggerated, pathologic response of the preterm infant to this transition. Anemia of prematurity is a normocytic, normochromic, hyporegenerative anemia characterized by a low serum erythropoietin (EPO) level, often despite a remarkably reduced hemoglobin concentration 4. Nutritional deficiencies of iron, vitamin E, vitamin B-12, and folate may exaggerate the degree of anemia, as may blood loss and/or a reduced red cell life span.

The anemia of prematurity is caused by untimely birth occurring before placental iron transport and fetal erythropoiesis are complete, by phlebotomy blood losses taken for laboratory testing, by low plasma levels of erythropoietin due to both diminished production and accelerated catabolism, by rapid body growth and need for commensurate increase in red cell volume/mass, and by disorders causing red blood cell losses due to bleeding and/or hemolysis 3.

The risk of anemia of prematurity is inversely related to gestational maturity and birthweight 4. As many as half of infants of less than 32 weeks gestation develop anemia of prematurity. Anemia of prematurity is not typically a significant issue for infants born beyond 32 weeks’ gestation.

Race and sex have no influence on the incidence of anemia of prematurity.

Testosterone is believed to be at least partially responsible for a slightly higher hemoglobin level in male infants at birth, but this effect is of no significance with regard to risk of anemia of prematurity. The nadir of the hemoglobin level is typically observed 4-10 weeks after birth in the tiniest infants, with hemoglobin levels of 8-10 g/dL if birthweight was 1200-1400 grams, or 6-9 g/dL at birth weights of less than 1200 grams and to approximately 7 g/dL in infants with birth weights <1 kg 4.

Anemia of prematurity is usually not serious. Anemia of prematurity spontaneously resolves in many premature infants within 3-6 months of birth 4. In others, however, medical intervention is required, because the low oxygen levels in a premature infant can make other problems worse, such as heart and lung problems. Most infants with birth weight <1.0 kg are given multiple red blood cell (RBC) transfusions within the first few weeks of life 3.

Red blood cell transfusions are the mainstay of therapy for anemia of prematurity with recombinant human erythropoietin (EPO) largely unused because it fails to substantially diminish red blood cell transfusion needs despite exerting substantial erythropoietic effects on neonatal marrow.

Figure 1. Blood composition

blood composition

blood composition

Footnote: Blood consists of a liquid portion called plasma and a solid portion (the formed elements) that includes red blood cells, white blood cells, and platelets. When blood components are separated by centrifugation, the white blood cells and platelets form a thin layer, called the “buffy coat,” between the plasma and the red blood cells, which accounts for about 1% of the total blood volume. Blood cells and platelets can be seen under a light microscope when a blood sample is smeared onto a glass slide.

Figure 2. Red blood cells (normal red blood cells)

red blood cells

Anemia of prematurity causes

The three basic mechanisms for the development of anemia of prematurity include:

  1. Inadequate red blood cell production,
  2. Shortened red blood cell life span,
  3. Blood loss.

Taken together, the premature infant is at risk for the development of anemia of prematurity because of limited red blood cell synthesis during rapid growth, a diminished red blood cell life span, and an increased loss of red blood cells.

Inadequate red blood cell production

The first mechanism of anemia is inadequate red blood cell production for the growing premature infant. The location of erythropoietin (EPO) and red blood cell production changes during gestation. Erythropoietin (EPO) synthesis initially occurs in the fetal liver but gradually shifts toward the kidney as gestation advances. By the end of gestation, however, the liver remains the major source of erythropoietin (EPO).

Fetal erythrocytes are produced in the yolk sac during the first few weeks of embryogenesis. The fetal liver becomes more important as gestation advances and, by the end of the first trimester, has become the primary site of erythropoiesis. Bone marrow then begins to take on a more active role in producing erythrocytes. By about 32 weeks’ gestation, the burden of erythrocyte production in the fetus is shared evenly by liver and bone marrow. By 40 weeks’ gestation, the marrow is the sole erythroid organ. Premature delivery does not accelerate the ontogeny of these processes.

Although erythropoietin (EPO) is not the only erythropoietic growth factor in the fetus, it is the most important. Erythropoietin (EPO) is synthesized in response to anemia and consequent relative tissue hypoxia. The degree of anemia and hypoxia required to stimulate erythropoietin (EPO) production is far greater for the fetal liver than for the fetal kidney. Erythropoietin (EPO) production may not be stimulated until a hemoglobin concentration of 6-7 g/dL is reached. As a result, new red blood cell production in the extremely premature infant, whose liver remains the major site of erythropoietin (EPO) production, is blunted despite what may be marked anemia. In addition, erythropoietin (EPO), whether endogenously produced or exogenously administered, has a larger volume of distribution and is more rapidly eliminated by neonates, resulting in a curtailed time for bone marrow stimulation.

Erythroid progenitors in premature infants are quite responsive to erythropoietin (EPO), but the response may be blunted if iron or other substrate or co-factor stores are insufficient. Another potential problem is that while the infant may respond appropriately to increased erythropoietin (EPO) concentrations with increased reticulocyte counts, rapid growth may prevent the appropriate increase in hemoglobin concentration.

Shortened red blood cell life span or hemolysis

Also important in the development of anemia of prematurity is that the average life span of a neonatal red blood cell is only one half to two thirds that of an adult red blood cell. Cells of the most immature infants may survive only 35-50 days. The shortened red blood cell life span of the neonate is a result of multiple factors, including diminished levels of intracellular adenosine triphosphate (ATP), carnitine, and enzyme activity; increased susceptibility to lipid peroxidation; and increased susceptibility of the cell membrane to fragmentation.

Blood loss

Finally, blood loss may contribute to the development of anemia of prematurity. If the neonate is held above the placenta for a time after delivery, fetal-placental transfer of blood may occur. Conversely, delayed cord clamping may lessen the degree of anemia of prematurity 5, although a study by Elimian et al 6 did not find this to be true. More commonly, because of the need to closely monitor the tiny infant, frequent samples of blood are removed for various tests. These losses are often 5-10% of the total blood volume.

Anemia of prematurity differential diagnoses

Conditions to consider in the differential diagnosis of anemia of prematurity are those which diminish red cell production, increase red cell destruction, or cause blood loss.

  • Acute Anemia
  • Birth Trauma
  • Chronic Anemia
  • Head Trauma
  • Hemolytic Disease of the Newborn
  • Parvovirus B19 Infection
  • Intraventricular Hemorrhage in the Preterm Infant

Conditions that diminish red blood cell synthesis are as follows:

  • Bone marrow infiltration
  • Bone marrow depression (eg, pancytopenia, drugs)
  • Diamond-Blackfan anemia
  • Substrate deficiencies (eg, iron, vitamin E, folic acid)
  • Congenital fetal infections (eg, cytomegalovirus, parvovirus, syphilis)

Conditions that cause hemolysis are as follows:

  • Congenital fetal infections (eg, cytomegalovirus, parvovirus, syphilis)
  • Acute systemic infections (leading to disseminated intravascular coagulation)
  • Abnormal red blood cells (spherocytosis, elliptocytosis)
  • Nonspherocytic hemolytic anemias (eg, G6PD deficiency, kinase and isomerase deficiencies)
  • Hemolytic disease of the newborn (Rh, ABO, other major blood-group incompatibilities between mother and fetus)

Conditions that reduce blood volume are as follows:

  • Twin-to-twin transfusion syndrome (donor twin)
  • Iatrogenic (eg, excessive blood sampling)
  • Hemorrhage (eg, gastrointestinal, central nervous system, subcutaneous tissues)

Anemia of prematurity symptoms

Many clinical findings have been attributed to anemia of prematurity, but they are neither specific nor diagnostic. These symptoms may include the following:

  • Poor weight gain despite adequate caloric intake
  • Cardiorespiratory symptoms such as tachycardia, tachypnea, and flow murmurs
  • Decreased activity, lethargy, and difficulty with oral feeding
  • Pallor
  • Increase in apneic and bradycardic episodes, and worsened periodic breathing
  • Metabolic acidemia – Increased lactic acid secondary to increased cellular anaerobic metabolism in relatively hypoxic tissues

Anemia of prematurity diagnosis

The following are useful laboratory studies:

  • Complete blood count (CBC) – White blood cell (WBC) and platelet values are normal in anemia of prematurity. Low hemoglobin values, below 10 g/dL, are found. They may descend to a nadir of 6-7 g/dL. Lowest levels are generally observed in the smallest infants. Red blood cell indices are normal (eg, normochromic, normocytic) for age.
  • Reticulocyte count – The reticulocyte count is low when the degree of anemia is considered, as a result of the low levels of erythropoietin (EPO). Conversely, an elevated reticulocyte count is not consistent with the diagnosis of anemia of prematurity.
  • Peripheral blood smear – Red blood cell morphology should be normal. Red blood cell precursors may appear to be more prominent.
  • Maternal and infant blood typing; direct antibody test (Coombs) – The direct Coombs test result may be coincidentally positive. Despite this, it is important to ensure an immune-mediated hemolytic process related to maternal-fetal blood group incompatibility (hemolytic disease of the newborn) is not present.
  • Serum bilirubin – An elevated serum bilirubin level should suggest other possible explanations for the anemia. These would include hemolytic entities, such as G-6-PD deficiency or other kinase/isomerase/enzyme deficiencies, or more common causes such as infection or hemolytic disease of the newborn.
  • Lactic acid – Elevated lactic acid levels have been suggested by some to be useful as an aid to determine the need for transfusion.

Anemia of prematurity treatment

Medical treatment options are blood transfusion(s), recombinant erythropoietin (EPO) treatment, and observation.

Observation may be the best course of action for infants who are asymptomatic, not acutely ill, and are receiving adequate nutrition. Adequate amounts of vitamin E, vitamin B-12, folate, and iron are important to blunt the expected decline in hemoglobin levels in the premature infant. Periodic measurements of the hematocrit level in infants with anemia of prematurity are necessary after hospital discharge. Once a steady increase in the hematocrit level has been established, only routine checks are required.

Packed red blood cell transfusions

Packed red blood cell transfusions are the mainstay of therapy for anemia of prematurity. The frequency of blood transfusion varies with gestational age, degree of illness, and, interestingly, the hospital evaluated. Unfortunately, there is considerable disagreement about the indication, timing, and efficacy of packed red blood cell transfusion.

Guidelines for transfusing red blood cells to preterm neonates are controversial, and practices vary greatly 7. This lack of a universal approach stems from limited knowledge of the cellular and molecular biology of erythropoiesis during the perinatal period, an incomplete understanding of infant physiological/adaptive responses to anemia, and contrary/controversial transfusion practice guidelines as based on results of randomized clinical trials and expert opinions. Generally, red blood cell transfusions are given to maintain a level of blood hemoglobin or hematocrit believed to be optimal for each neonate’s clinical condition. Guidelines for red blood cell transfusions, judged to be reasonable by most neonatologists to treat the anaemia of prematurity, are listed by Table 1. These guidelines are very general, and it is important that terms such as “severe” and “symptomatic” be defined to fit local transfusion practices/policies. Importantly, guidelines are not mandates for red blood cell transfusions that must be followed; they simply suggest situations when an red blood cell transfusion would be judged to be reasonable/acceptable.

The decision to give a transfusion should not be made lightly, because significant infectious, hematologic, immunologic, and metabolic complications are possible. Late-onset necrotizing enterocolitis has been reported in stable-growing premature infants electively transfused for anemia of prematurity 8. Transfusions also transiently decrease erythropoiesis and EPO levels. There is also agreement that the number of transfusions, as well as the number of donor exposures, should be reduced as much as possible.

Clinical trials designed to determine the efficacy of blood transfusions in relieving symptoms ascribed to anemia of prematurity have produced conflicting results 9. Improved growth has been an inconsistent finding. While some studies have demonstrated a decrease in apneic episodes after blood transfusion, others have found similar results with simple crystalloid volume expansion.

Subjective improvement in activity, decreased lethargy, and improved feeding have been described in some studies. Blood transfusions have been documented to decrease lactic acid levels in otherwise healthy preterm infants who are anemic. Blood transfusions have reduced tachycardia in anemic infants who are transfused.

Some medical professionals have suggested using lactate levels as an aid in determining the need for transfusion.

Table 1. Allogeneic red blood cell transfusions for the anemia of prematurity

Transfuse to maintain the blood hematocrit per each clinical situation:

  • > 40% (35 to 45% *) for severe cardiopulmonary disease
  • > 30% for moderate cardiopulmonary disease
  • > 30% for major surgery
  • >25% (20 to 25% *) for symptomatic anemia
  • > 20% for asymptomatic anemia

*Reflects practices that vary among neonatologists. Thus, any value within range is acceptable for local practices.

Reducing the number of transfusions

Studies from individual centers have documented a marked decrease in the administration of packed red blood cell transfusions in the past decades, even before the use of EPO became more frequent. This decrease in transfusions is almost certainly multifactorial in origin. Adoption of standardized transfusion protocols that take various factors into account, including hemoglobin levels, degree of cardiorespiratory disease, and traditional signs and symptoms of pathologic anemia, are acknowledged as an important factor in this reduction. A restricted transfusion protocol may decrease the number of transfusions while also decreasing the hematocrit at discharge 10.

A 2011 study 11 evaluated 41 preterm infants with birth weights of 500-1300 g who were enrolled in a clinical trial that compared high and low hematocrit thresholds for transfusion. A rise in systemic oxygen transport and a decrease in lactic acid after transfusion was noted in both groups; however, oxygen consumption did not change significantly in either group. In the low hematocrit group only, cardiac output and fractional oxygen extraction fell after transfusion, which shows that these infants had increased their cardiac output to maintain adequate tissue oxygen delivery in response to anemia. The results demonstrate that infants with low hematocrit thresholds may benefit from transfusion, while transfusion in those with high hematocrit thresholds may provide no acute physiological benefit 11.

The Premature Infant in Need of Transfusion study 12 showed that transfusing infants to maintain higher hemoglobin level (8.5-13.5 g/dL) conferred no benefit in terms of mortality, severe morbidity, or apnea intervention compared with infants transfused to maintain a low hemoglobin levels (7.5-11.5 g/dL).

These findings differ from the Iowa study, which found less parenchymal brain hemorrhage, periventricular leukomalacia, and apnea in infants whose transfusion criteria was not restricted and whose hemoglobin level was higher. Clearly, no universally accepted guidelines for transfusion in infants with anemia of prematurity are available at this time, and clinicians must determine their individual standardized transfusion practices.

Anemia of prematurity guidelines

No universally accepted guidelines for transfusion in infants with anemia of prematurity are available at this time, and clinicians must determine their individual standardized transfusion practices.

As an example, note the Children’s Hospital of Wisconsin Transfusion Committee guidelines for consideration:

  • An infant with a hemoglobin (Hb) level of less than 8 g/dL may be transfused at the discretion of the attending physician
  • A stable infant with a hemoglobin level of 8-10 g/dL without clinical symptoms or other exceptions listed below may be transfused
  • An infant with a hemoglobin level of 11-13 g/dL without a supplemental oxygen or continuous positive airway pressure (CPAP) requirement, apnea/bradycardia, significant tachycardia or tachypnea, or other exceptions listed below should not be transfused
  • An infant with a hemoglobin level of more than 13 g/dL without an oxygen requirement of more than 40% by hood, CPAP, or ventilator; hypotension that requires pressor medication; major surgery; or other exceptions listed below should not be transfused
  • An infant with a hemoglobin level of more than 15 g/dL without cyanotic heart disease, extracorporeal membrane oxygenation (ECMO) therapy, regional oxygen saturations less than 50%, or hypotension that requires pressor medications should not be transfused
  • An infant with a history of massive blood loss may be transfused at the discretion of the attending physician

It is of obvious importance to discuss with the family their child’s need for transfusion and to obtain consent before the transfusion. It is also important to discuss with parents the normal course of anemia, the criteria for and risks associated with transfusions, and the advantages and disadvantages of erythropoietin (EPO) administration. Also necessary is consideration of the family’s religious beliefs regarding transfusions.

Reducing the number of donor exposures

Reducing the number of donor exposures is also important. Preservatives and additive systems allow blood to be stored safely for as long as 35-42 days. This can be accomplished by using packed red blood cells stored in preservatives (eg, citrate-phosphate-dextrose-adenine [CPDA-1]) and additive systems (eg, Adsol). Infants may be assigned a specific unit of blood, which may suffice for treatment during their entire hospitalization and thus limit exposure to the single donor of that unit. Concerns that stored blood might increase serum potassium levels are unfounded if the transfused volume is low.

Complications

Potential complications of transfusion include the following:

  • Infection (eg, hepatitis, cytomegalovirus [CMV], human immunodeficiency virus [HIV], syphilis)
  • Fluid overload and electrolyte imbalances
  • Exposure to plasticizers
  • Hemolysis
  • Posttransfusion graft versus host disease

An important tool in reducing at least one of these transfusion risks is to use all available screening techniques for infection. The risk of cytomegalovirus (CMV) transmission can be dramatically reduced by use of CMV-safe blood. This can be accomplished by using CMV serology–negative cells, along with blood processed through leukocyte-reduction filters or inverted spin technique. These methods also reduce other WBC-associated infectious agents (eg, Epstein-Barr virus, retroviruses, Yersinia enterocolitica) by yielding a leukocyte-poor suspension of packed red blood cells. The American Red Cross now provides exclusively leukocyte-reduced blood to hospitals in the United States.

Recombinant Erythropoietin treatment

Multiple investigations have established that premature infants respond to exogenously administered recombinant human EPO and supplemental iron with a brisk reticulocytosis. Subcutaneous administration of EPO may be preferred, as intravenous administration has increased urinary losses. Although EPO cannot prevent early transfusions, modest decreases in the frequency of late packed red blood cell transfusions have been documented. Additional iron supplementation is necessary during exogenous EPO treatment.

Trials have evaluated the impact of EPO treatment in populations of the most immature neonates. These studies likewise have demonstrated that infants with very low birth weight (VLBW) are capable of responding to EPO with a reticulocytosis.

Studies and a Cochrane Neonatal Systemic review suggest an association between exogenous EPO administration and retinopathy of prematurity 13.

Yasmeen et al 14 studied 60 preterm low birth weight infants and concluded that short-term recombinant human erythropoietin with iron and folic acid was effective in preventing anemia of prematurity.

EPO with iron does not adversely affect growth or developmental outcomes, but the impact on the number of transfusions a premature infant receives ranges from nonexistent to small.

At this time, no agreement regarding the safety, timing, dosing, route, or duration of therapy has been established. In short, the cost-benefit ratio for EPO has yet to be clearly established, and this medication is not universally accepted as a standard therapy for an infant with anemia of prematurity.

Anemia of prematurity prognosis

Spontaneous recovery of mild anemia of prematurity may occur 3-6 months after birth. In more severe, symptomatic cases, medical intervention may be required.

References
  1. Martin JA, Hamilton BE, Sutton PD, et al. Births: final data for 2008 national Vital Statistics Reports. Centers for Disease Control and Prevention. 2009;57:7.
  2. Maier RJ, Sonntag J, Walka MM, et al. Changing practices of red blood cell transfusions in infants with birth weights less than 1000 g. J Pediatr. 2000;136:220–224.
  3. Strauss RG. Anaemia of prematurity: pathophysiology and treatment. Blood Rev. 2010;24(6):221-225. doi:10.1016/j.blre.2010.08.001 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2981681
  4. Anemia of Prematurity. https://emedicine.medscape.com/article/978238-overview
  5. Ultee CA, van der Deure J, Swart J, Lasham C, van Baar AL. Delayed cord clamping in preterm infants delivered at 34 36 weeks’ gestation: a randomised controlled trial. Arch Dis Child Fetal Neonatal Ed. 2008 Jan. 93(1):F20-3.
  6. Elimian A, Goodman J, Escobedo M, Nightingale L, Knudtson E, Williams M. Immediate compared with delayed cord clamping in the preterm neonate: a randomized controlled trial. Obstet Gynecol. 2014 Dec. 124 (6):1075-9.
  7. dos Santos AMN, Guinsburg R, Procianoy RS, dos SR, Sadeck L, Netto AA, Rugolo LM, et al. Variability on red blood cell transfusion practices among Brazilian neonatal intensive care units. Transfusion. 2010;50:150–159.
  8. Singh R, Shah BL, Frantz ID 3rd. Necrotizing enterocolitis and the role of anemia of prematurity. Semin Perinatol. 2012 Aug. 36(4):277-82.
  9. Bell EF, Nahmias C, Sinclair JC, Zipursky A. Changes in circulating red cell volume during the first 6 weeks of life in very-low-birth-weight infants. Pediatr Res. 2014 Jan. 75 (1-1):81-4.
  10. Bell EF, Strauss RG, Widness JA, et al. Randomized trial of liberal versus restrictive guidelines for red blood cell transfusion in preterm infants. Pediatrics. 2005 Jun. 115(6):1685-91.
  11. Fredrickson LK, Bell EF, Cress GA, et al. Acute physiological effects of packed red blood cell transfusion in preterm infants with different degrees of anaemia. Arch Dis Child Fetal Neonatal Ed. 2011 Jul. 96(4):F249-53.
  12. Kirpalani H, Whyte RK, Andersen C, et al. The Premature Infants in Need of Transfusion (PINT) study: a randomized, controlled trial of a restrictive (low) versus liberal (high) transfusion threshold for extremely low birth weight infants. J Pediatr. 2006 Sep. 149(3):301-307.
  13. Suk KK, Dunbar JA, Liu A, et al. Human recombinant erythropoietin and the incidence of retinopathy of prematurity: a multiple regression model. J AAPOS. 2008 Jun. 12(3):233-8.
  14. Yasmeen BH, Chowdhury MA, Hoque MM, Hossain MM, Jahan R, Akhtar S. Effect of short-term recombinant human erythropoietin therapy in the prevention of anemia of prematurity in very low birth weight neonates. Bangladesh Med Res Counc Bull. 2012 Dec. 38(3):119-23.
read more
Diet, Food & FitnessDrugs & SupplementsFoodsImmune and Hematology SystemNatural RemediesSupplements

L glutamine

L-Glutamine

What is L-Glutamine

L-glutamine is in a class of medications called amino acids. It works by helping to prevent damage to red blood cells. L-glutamine is a non-essential branched-chain amino acid that is present abundantly throughout the body and is involved in many metabolic processes 1. L-glutamine is an important non-toxic nitrogen carrier in the body and an essential component of diet, especially dairy products, fish and green leafy vegetables 2. L-glutamine participates in a variety of physiological functions, and is a major fuel source of enterocytes (intestine or gut cells) and is a substrate for gluconeogenesis (glucose formation) in the kidney, lymphocytes (white blood cells) and monocytes (white blood cells). L-glutamine is also a nutrient in muscle protein metabolism in response to infection, inflammation and muscle trauma 3. Because of glutamine’s importance as a nitrogen carrier and respiratory fuel for enterocytes of the gut and other rapidly proliferating cells, including lymphocytes and fibroblasts, glutamine can be considered as a conditionally essential amino acid 4. Although there are no known drug interactions with glutamine, physiological antagonism may occur with lactulose when given to treat high ammonia levels in liver failures. In some patients, glutamate may lead to brain excitation, and in patients with seizure, may make the drug less effective.

L-glutamine is used by doctors as prescription medicine to reduce the frequency of painful episodes (crises) in adults and children 5 years of age and older with sickle cell anemia (an inherited blood disorder in which the red blood cells are abnormally shaped [shaped like a sickle] and cannot bring enough oxygen to all parts of the body) 5. And L-glutamine is often prescribed to treat short bowel syndrome (short gut syndrome). Glutamine is used together with human growth hormone and a specialized diet to treat short bowel syndrome. This medicine is available only with your doctor’s prescription.

  • L-glutamine has Pregnancy Category C: Animal studies have shown an adverse effect and there are no adequate studies in pregnant women OR no animal studies have been conducted and there are no adequate studies in pregnant women.
  • Breast Feeding: There are no adequate studies in women for determining infant risk when using this medication during breastfeeding. Weigh the potential benefits against the potential risks before taking this medication while breastfeeding.

In addition to L-glutamine usage in sickle cell anemia and short bowel syndrome, L-glutamine supplement may have an important role in the prevention of gastrointestinal, neurologic and, possibly, cardiac complications of cancer therapy 2. The influence of glutamine on body homeostasis is protean (able to do many different things, versatile). States of physiologic stress, including those resulting from the treatment of malignant disease, are characterized by a relative deficiency of glutamine. These complications often negatively affect the quality of life and may also lead to changes in therapy, which potentially alter efficacy. L-glutamine may also improve the therapeutic index of both chemotherapy and radiation, increasing cytotoxicity while concurrently protecting against toxicity. However, the current evidence is not sufficient to recommend its regular use. Further studies of glutamine supplementation in these areas is warranted and multicentric, placebo-controlled phase III studies are needed to evaluate the role of L-glutamine for the prevention of mucositis, neurotoxicity and cardiotoxicity, and for the prevention of hepatic venoocclusive disease in patients undergoing hematopoietic cell transplantation before any definitive recommendation can be made.

Figure 1. L-Glutamine

L-Glutamine

 

What does L-glutamine do?

Glutamine is a major component of tissue of the skeletal muscle, which is the main site for the synthesis and storage of L-glutamine. When the supply of glutamine in plasma is inadequate to meet the demand, glutamine synthesis occurs in skeletal muscle and liver. Glutamine is transported to the neurons and, by the enzyme glutaminase, is converted to glutamate – the potential excitotoxin. L-glutamine accounts for 30–35% of the amino acid nitrogen in the plasma. It contains two ammonia groups, one from its precursor, glutamate, and the other from free ammonia in the bloodstream. Glutamine plays an important role to prevent fluctuations in the levels of ammonia in blood by acting as a “nitrogen shuttle.” It does so by acting as a buffer, accepting and then releasing excess ammonia when needed to form other amino acids, amino sugars, nucleotides and urea. This capacity to accept and donate nitrogen makes glutamine the major vehicle for nitrogen transfer among tissues. Glutamine is one of the three amino acids involved in glutathione synthesis. Glutathione synthesis, an important intracellular antioxidant and hepatic detoxifier, is comprised of glutamic acid, cysteine and glycine 6. Glutamine is one of the most important substrates for ammoniagenesis in the gut and the kidney due to its important role in the regulation of acid–base homeostasis 7. It decomposes readily to yield ammonia and glutamate or via intramolecular catalysis to pyroglutamate. Deamidation of glutamine via the enzyme glutaminase produces glutamate, a precursor of gamma-amino butyric acid. The transfer of the amide nitrogen from glutamine via the amido transferase reaction is involved in the biosynthesis of purines and pyrimidines and in the production of hexosamines. Glutamine via glutamate is converted to α-ketoglutarate, an integral component of the citric acid cycle. It is a component of the antioxidant glutathione synthesis and of the polyglutamated folic acid. The cyclization of glutamate produces proline, an amino acid important for the synthesis of collagen and connective tissue. However, excess glutamine in a protein is of pathological importance, and a number of neurodegenerative diseases have been found to be due to a CAG expansion that causes expansion of glutamine repeats in affected proteins (CAA and CAG codons are responsible for the insertion of glutamine from its transfer RNA with its anti-codon triplet into the genetically determined position of the coded polypeptide chain). This leads to abnormal protein folding 8 and neuronal diseases 9.

Glutamine is involved in many metabolic processes in the body. Glutamine is converted to glucose when more glucose is required by the body as an energy source. Glutamine also plays a part in maintaining proper blood glucose levels and the right pH range. It is also used by white blood cells and is important for immune function. Glutamine assists in maintaining the proper acid/alkaline balance in the body, and is the basis of the building blocks for the synthesis of RNA and deoxyribonucleic acid (DNA). Glutamine regulates the expression of certain genes, including those that govern certain protective enzymes, and helps regulate the biosynthesis of DNA and RNA. Construction of DNA is dependent on adequate amounts of glutamine. It also increases the body’s ability to secrete human growth hormone, which assists in metabolizing body fat and helps to support new muscle tissue growth. The glutamic acid–glutamine interconversion is of central importance to the regulation of the levels of toxic ammonia in the body, and thus among all the amino acids of blood plasma, glutamine has the highest concentration.

L-glutamine benefits

Glutamine is one of the most common plasma amino acids, and its concentration often decreases post-operatively 10, during sepsis 11 and after multiple trauma 12 or major burns 13, similar to a fall in the concentrations of many other amino acids, electrolytes, minerals and trace elements; therefore, it seems prudent to give glutamine supplementation in all these conditions 14.

Short Bowel Syndrome

Short bowel syndrome is a group of problems related to poor absorption of nutrients. Short bowel syndrome typically occurs in people who have:

  • had at least half of their small intestine removed and sometimes all or part of their large intestine removed
  • significant damage of the small intestine
  • poor motility, or movement, inside the intestines

Short bowel syndrome may be mild, moderate, or severe, depending on how well the small intestine is working.

People with short bowel syndrome cannot absorb enough water, vitamins, minerals, protein, fat, calories, and other nutrients from food. What nutrients the small intestine has trouble absorbing depends on which section of the small intestine has been damaged or removed.

What causes Short Bowel Syndrome ?

The main cause of short bowel syndrome is surgery to remove a portion of the small intestine. This surgery can treat intestinal diseases, injuries, or birth defects.

Some children are born with an abnormally short small intestine or with part of their bowel missing, which can cause short bowel syndrome. In infants, short bowel syndrome most commonly occurs following surgery to treat necrotizing enterocolitis, a condition in which part of the tissue in the intestines is destroyed 15.

Short bowel syndrome may also occur following surgery to treat conditions such as:

  • cancer and damage to the intestines caused by cancer treatment
  • Crohn’s disease, a disorder that causes inflammation, or swelling, and irritation of any part of the digestive tract
  • gastroschisis, which occurs when the intestines stick out of the body through one side of the umbilical cord
  • internal hernia, which occurs when the small intestine is displaced into pockets in the abdominal lining
  • intestinal atresia, which occurs when a part of the intestines doesn’t form completely
  • intestinal injury from loss of blood flow due to a blocked blood vessel
  • intestinal injury from trauma
  • intussusception, in which one section of either the large or small intestine folds into itself, much like a collapsible telescope
  • meconium ileus, which occurs when the meconium, a newborn’s first stool, is thicker and stickier than normal and blocks the ileum
  • midgut volvulus, which occurs when blood supply to the middle of the small intestine is completely cut off
  • omphalocele, which occurs when the intestines, liver, or other organs stick out through the navel, or belly button

Even if a person does not have surgery, disease or injury can damage the small intestine.

How common is Short Bowel Syndrome ?

Short bowel syndrome is a rare condition. Each year, short bowel syndrome affects about three out of every million people 15.

What are the signs and symptoms of Short Bowel Syndrome ?

The main symptom of short bowel syndrome is diarrhea—loose, watery stools. Diarrhea can lead to dehydration, malnutrition, and weight loss. Dehydration means the body lacks enough fluid and electrolytes—chemicals in salts, including sodium, potassium, and chloride—to work properly. Malnutrition is a condition that develops when the body does not get the right amount of vitamins, minerals, and nutrients it needs to maintain healthy tissues and organ function. Loose stools contain more fluid and electrolytes than solid stools. These problems can be severe and can be life threatening without proper treatment.

Other signs and symptoms may include:

  • bloating
  • cramping
  • fatigue, or feeling tired
  • foul-smelling stool
  • heartburn
  • too much gas
  • vomiting
  • weakness

People with short bowel syndrome are also more likely to develop food allergies and sensitivities, such as lactose intolerance. Lactose intolerance is a condition in which people have digestive symptoms—such as bloating, diarrhea, and gas—after eating or drinking milk or milk products.

What are the complications of Short Bowel Syndrome ?

The complications of short bowel syndrome may include

  • malnutrition
  • peptic ulcers—sores on the lining of the stomach or duodenum caused by too much gastric acid
  • kidney stones—solid pieces of material that form in the kidneys
  • small intestinal bacterial overgrowth—a condition in which abnormally large numbers of bacteria grow in the small intestine

How is Short Bowel Syndrome treated ?

A health care provider will recommend treatment for short bowel syndrome based on a patient’s nutritional needs. Treatment may include

  • nutritional support
  • medications
  • surgery
  • intestinal transplant

Nutritional Support

The main treatment for short bowel syndrome is nutritional support, which may include the following:

  • Oral rehydration. Adults should drink water, sports drinks, sodas without caffeine, and salty broths. Children should drink oral rehydration solutions—special drinks that contain salts and minerals to prevent dehydration—such as Pedialyte, Naturalyte, Infalyte, and CeraLyte, which are sold in most grocery stores and drugstores.
  • Parenteral nutrition. This treatment delivers fluids, electrolytes, and liquid vitamins and minerals into the bloodstream through an intravenous (IV) tube—a tube placed into a vein. Health care providers give parenteral nutrition to people who cannot or should not get their nutrition or enough fluids through eating.
  • Enteral nutrition. This treatment delivers liquid food to the stomach or small intestine through a feeding tube—a small, soft, plastic tube placed through the nose or mouth into the stomach. Gallstones—small, pebble like substances that develop in the gallbladder—are a complication of enteral nutrition.
  • Vitamin and mineral supplements. A person may need to take vitamin and mineral supplements during or after parenteral or enteral nutrition.
  • Special diet. A health care provider can recommend a specific diet plan for the patient that may include:
    • small, frequent feedings
    • avoiding foods that can cause diarrhea, such as foods high in sugar, protein, and fiber
    • avoiding high-fat foods.

Bone marrow transplant

Bone marrow transplant is a sophisticated procedure consisting of the administration of high-dose chemoradiotherapy followed by intravenous infusion of hemopoietic stem cells to re-establish marrow function when the bone marrow is damaged or defective. Bone marrow transplant is used in the treatment of solid tumors, hematological diseases and autoimmune disorders. Glutamine has protein-anabolic effects and has shown a clear reduction of complications in patients undergoing bone marrow transplant who exhibit post-transplant body protein wasting, gut mucosal injury leading to mucositis of gastrointestinal tract, acute graft versus host disease and immunodeficiency. Studies indicate that enteral and parenteral glutamine supplementation is well tolerated and potentially efficacious after high-dose chemotherapy or bone marrow transplant for cancer treatment. Although not all studies demonstrate benefits, sufficient data has been published to suggest that this nutrient should be considered as adjunctive metabolic support of some individuals undergoing marrow transplant 16. However, bone marrow transplant is a rapidly evolving clinical procedure with regard to the conditioning and supportive protocols used. Thus, additional randomized, double-blind, controlled clinical trials are indicated to define the efficacy of glutamine with current bone marrow transplant regimens 17.

Glutamine and Cancer

Numerous studies on glutamine metabolism in cancer indicate that many tumors are avid glutamine consumers in vivo and in vitro. As a consequence of progressive tumor growth, host glutamine depletion develops and becomes a hallmark. This glutamine depletion occurs in part because the tumor behaves as a “glutamine trap” and also because of cytokine-mediated alterations in glutamine metabolism in host tissues. Animal and human studies that have investigated the use of glutamine-supplemented nutrition in the host with cancer suggest that pharmacologic doses of dietary glutamine may be beneficial. Understanding the control of glutamine metabolism in the tumor-bearing host not only improves the knowledge of metabolic regulation in the patient with cancer but also leads to improved nutritional support regimens targeted to benefit the host.

Glutamine supplementation in chemotherapy

The results of glutamine supplementation and oncology in animals and humans are conflicting 18. In vitro (test tube) studies reveal an increase in cellular growth with glutamine supplementation 19. While in vivo (animal) studies show the opposite effect, i.e. reduction in tumor growth 20. Glutamine uptake in patients with colon cancer, regardless of tumor size and cell type, is comparable to uptake in patients with healthy intestinal tissue 21, also enteral diet containing glutamine increase muscle glutamine in rats by 60% without increasing tumor growth or tumor glutamine use 22. Glutamine supplementation in rats receiving methotrexate chemotherapy causes reduction in methotrexate-induced side-effects, including mucositis, and improved survival is observed 23. Mucosal ulceration in rats subjected to abdominal radiation is also prevented 24.

GLUTAMINE: ROLE IN INCREASING SELECTIVITY OF CHEMOTHERAPEUTIC AGENTS

Chemotherapy doses are limited by toxicity to normal tissues. Intravenous glutamine protects liver cells from oxidant injury by increasing intracellular glutathione synthesis content 25. The effects of oral glutamine on tumor and host glutathione synthesis metabolism and response to methotrexate have been studied in rat models of sarcoma as well as in human patients with inflammatory breast cancer. Feeding the glutamine-enriched diets to rats receiving methotrexate decreases tumor glutathione synthesis while increasing or maintaining host glutathione synthesis stores 26. Diminished glutathione synthesis levels in tumor cells increases susceptibility to chemotherapy. Significantly decreased glutathione synthesis content in tumor cells in the glutamine-supplemented group correlates with enhanced tumor volume loss 27. These data suggest that oral glutamine supplementation will enhance the selectivity of antitumor drugs by protecting normal tissues from and possibly sensitizing tumor cells to radiation-induced and chemotherapy treatment-related injury 28.

GLYCYL-GLUTAMINE-DIPEPTIDE IN THE PARENTERAL NUTRITION OF PATIENTS WITH ACUTE LEUKEMIA UNDERGOING INTENSIVE CHEMOTHERAPY

The effects of parenteral glycyl-glutamine supplementation in patients with acute leukemia receiving intensive conventional chemotherapy was evaluated in a randomized, double-blind, controlled study that compared a standard glutamine-free parenteral nutrition with a glycyl-glutamine-supplemented parenteral nutrition containing 20 g of glutamine. There was significant faster neutrophil recovery in the group that received glutamine supplementation along with high-dose cytarabine chemotherapy as compared with those patients receiving cytarabine regimen alone. There was no significant difference in the recovery of CD4+ or CD8+ lymphocytes or monocyte activation between the two groups. The authors concluded that there is a possible role of glutamine in the stimulation of lymphocyte proliferation 29.

Other Uses of L-glutamine

Possibly Effective for:

  • Burns. Administering glutamine through a feeding tube or intravenously (by IV) seems to reduce infections, shorten hospital stays, and improve wound healing in people with burns.
  • Critical illness (trauma). There is some evidence that glutamine keeps bacteria from moving out of the intestine and infecting other parts of the body after major injuries. However, not all evidence is consistent. It is not clear if glutamine reduces the risk of death in critically ill people. Some studies suggest that it might reduce the risk of death, while others do not.
  • Treating weight loss and intestinal problems in people with HIV/AIDs disease. Taking glutamine by mouth seems to help HIV/AIDS patients absorb food better and gain weight. Doses of 40 grams per day seem to produce the best effect.
  • Soreness and swelling inside the mouth, caused by chemotherapy treatments. Some evidence suggests that glutamine reduces soreness and swelling inside the mouth caused by chemotherapy. However, glutamine does not seem to have this effect for all chemotherapy patients. It is not clear which patients are likely to benefit. Some researchers suspect that chemotherapy patients who do not have enough glutamine to start with are most likely to be helped.
  • Surgery. Giving glutamine intravenously (by IV) along with intravenous nutrition seems to improve immune function and reduce complications related to infections after major surgery. Also, giving glutamine intravenously (by IV) along with intravenous nutrition after a bone marrow transplant seems to reduce the risk of infection and improve recovery compared to intravenous nutrition alone. However, not all people who undergo major surgery or who receive bone marrow transplants seem to benefit from glutamine.

Possibly Ineffective for 30:

  • Athletic performance. Taking glutamine by mouth does not seem to improve athletic performance.
  • Crohn’s disease. Taking glutamine by mouth does not seem to improve symptoms of Crohn’s disease.
  • Inherited disease that causes stones in the kidneys or bladder (Cystinuria). Taking glutamine by mouth does not seem to improve an inherited condition that causes stones to form in the kidneys or bladder.
  • Muscular dystrophy. Research shows that taking glutamine by mouth does not improve muscle strength in children with muscular dystrophy.

Insufficient Evidence for 30:

  • Diarrhea caused by drugs used to treat HIV. Early research shows that taking glutamine by mouth reduces the severity of diarrhea in people with HIV who are taking the drug nelfinavir.
  • Diarrhea caused by chemotherapy treatments. There is some evidence that glutamine might help to prevent diarrhea after chemotherapy, but not all research findings agree.
  • Reducing damage to the immune system during cancer treatment. There is some evidence that glutamine reduces damage to the immune system caused by chemotherapy. However, not all research findings agree.
  • Diarrhea. There are inconsistent findings about the effects of glutamine when used to treat diarrhea in children and infants. One early study suggests that taking glutamine by mouth reduces the duration of diarrhea in children. However, taking glutamine by mouth along with conventional rehydration solutions does not appear to have an advantage over rehydration solutions alone.
  • Low birth weight. There are inconsistent findings about the effects of glutamine in infants with low to very low birth weight. Some research suggests that using glutamine in feeding tubes decreases infections in some low birth weight infants. However, most research suggests that it does not benefit low birth weight infants.
  • Muscle and joint pains caused by the drug paclitaxel (Taxol, used to treat cancer). There is some evidence that glutamine might help to reduce muscle and joint pains caused by paclitaxel.
  • Inflammation of the pancreas (pancreatitis). An early study shows that giving glutamine intravenously (by IV) along with intravenous nutrition improves immune function but does not reduce the risk for complications or the amount of time spent in the hospital in people with pancreatitis.
  • Nutrition problems after major gut surgery (short bowel syndrome). Researchers have studied whether glutamine combined with growth hormone is effective in treating short bowel syndrome. This combination seems to help some patients become less dependent on tube feeding. However, glutamine alone does not seem to be effective.
  • Depression.
  • Moodiness.
  • Irritability.
  • Anxiety.
  • Attention deficit-hyperactivity disorder (ADHD).
  • Insomnia.
  • Stomach ulcers.
  • Ulcerative colitis.
  • Sickle cell anemia.
  • Treating alcoholism.
  • Other conditions.

More evidence is needed to rate glutamine for these uses.

When to take L-glutamine supplement

L-glutamine comes as a powder to be mixed with a liquid or soft wet food and taken by mouth twice a day. Take L-glutamine at around the same times every day. Follow the directions on your prescription label carefully, and ask your doctor or pharmacist to explain any part you do not understand. Take L-glutamine exactly as directed. Do not take more or less of it or take it more often than prescribed by your doctor.

You will need to mix the medication powder with 8 ounces (240 ml) of a liquid such as water, milk, or apple juice, or 4 to 6 ounces (120 to 180 ml) of a soft wet food such as applesauce or yogurt right before you take it. The liquid or food must be cold or room temperature. The powder does not need to be completely dissolved in the liquid or food before you take the mixture.

What special precautions should I follow when taking L-glutamine ?

Before taking L-glutamine:

  • tell your doctor and pharmacist if you are allergic to L-glutamine, any other medications.
  • tell your doctor and pharmacist what prescription and nonprescription medications, vitamins, nutritional supplements, and herbal products you are taking or plan to take. Your doctor may need to change the doses of your medications or monitor you carefully for side effects.
  • tell your doctor if you are pregnant, plan to become pregnant, or are breastfeeding. If you become pregnant while taking L-glutamine, call your doctor.

What special dietary instructions should I follow when taking L-glutamine ?

Unless your doctor tells you otherwise, continue your normal diet.

What should I do if I forget a dose ?

Take the missed dose as soon as you remember it. However, if it is almost time for the next dose, skip the missed dose and continue your regular dosing schedule. Do not take a double dose to make up for a missed one.

L-glutamine dosage

Your doctor will tell you how much L-glutamine medicine to use. Take this medicine exactly as directed by your doctor. Do not take more of it, do not take it more often, and do not take it for a longer time than your doctor ordered.

For patients using the oral powder for solution:

  • Mix a packet of this medicine with water just before using it.
  • Take it with a meal or snack every 2 to 3 hours while you are awake. Be sure to drink all of the mixture.
  • Do not use the medicine during the night unless your doctor tells you to.

For patients using the oral powder:

  • Mix the oral powder with 4 to 6 ounces (oz) of food (eg, applesauce, yogurt) or 8 oz of cold or room temperature beverage (eg, water, milk, or apple juice).
  • Complete dissolution of the mixture is not required.
  • Be sure to drink or swallow all of the mixture.

Dosing

The dose of this medicine will be different for different patients. Follow your doctor’s orders or the directions on the label. The following information includes only the average doses of this medicine. If your dose is different, do not change it unless your doctor tells you to do so.

The amount of medicine that you take depends on the strength of the medicine. Also, the number of doses you take each day, the time allowed between doses, and the length of time you take the medicine depend on the medical problem for which you are using the medicine.

  • For oral dosage form (powder for oral solution):

    • For short bowel syndrome:

      • Adults—30 grams (g) per day in divided doses (5 g taken 6 times a day) for up to 16 weeks. Taken with meals or snacks, 2 to 3 hours apart while awake. Be sure to drink all of the mixture. Do not use this medicine during the night unless your doctor tells you to.
      • Children—Use and dose must be determined by your doctor.
  • For oral dosage form (oral powder):

    • For sickle cell disease:

      • Adults and children 5 years of age and older and weighs greater than 65 kilograms (kg)—15 grams (g) per dose (3 packets per dose) 2 times a day or 30 g per day (6 packets per day).
      • Adults and children 5 years of age and older and weighs 30 to 65 kg—10 g per dose (2 packets per dose) 2 times a day or 20 g per day (4 packets per day).
      • Children 5 years of age and older and weighs less than 30 kg—5 g per dose (1 packet per dose) 2 times a day or 10 g per day (2 packets per day).
      • Children younger than 5 years of age—Use and dose must be determined by your doctor.
      • Mix the oral powder with 4 to 6 ounces (oz) of food (including apple sauce or yogurt) or with 8 oz of cold or room temperature beverage (including water, milk, or apple juice) as instructed by your doctor or pharmacist. Be sure to drink or swallow all of the mixture. Complete dissolution of the mixture is not required.

Missed dose: Take a dose as soon as you remember. If it is almost time for your next dose, wait until then and take a regular dose. Do not take extra medicine to make up for a missed dose.

Storage

Store the medicine in a closed container at room temperature, away from heat, moisture, and direct light. Keep from freezing.

Keep out of the reach of children.

Do not keep outdated medicine or medicine no longer needed.

Ask your healthcare professional how you should dispose of any medicine you do not use.

L-glutamine side effects and safety

Special Precautions & Warnings 31:

Children: Glutamine is POSSBILY SAFE when taken by mouth appropriately. Children aged 3 to 18 years should not be given doses that are larger than 0.7 grams per kg of weight daily. Not enough information is known about the safety of higher doses in children.

Pregnancy and breast-feeding: Not enough is known about the use of glutamine during pregnancy and breast-feeding. Stay on the safe side and avoid use.

Cirrhosis: Glutamine could make this condition worse. People with this condition should avoid glutamine supplements.

Severe liver disease with difficulty thinking or confusion (hepatic encephalopathy): Glutamine could make this condition worse. Do not use it.

Monosodium glutamate (MSG) sensitivity (also known as “Chinese restaurant syndrome”): If you are sensitive to MSG, you might also be sensitive to glutamine, because the body converts glutamine to glutamate.

Mania, a mental disorder: Glutamine might cause some mental changes in people with mania. Avoid use.

Seizures: There is some concern that glutamine might increase the likelihood of seizures in some people. Avoid use.

Drug Interactions

Although certain medicines should not be used together at all, in other cases two different medicines may be used together even if an interaction might occur. In these cases, your doctor may want to change the dose, or other precautions may be necessary. Tell your healthcare professional if you are taking any other prescription or nonprescription (over-the-counter [OTC]) medicine.

Other Interactions

Certain medicines should not be used at or around the time of eating food or eating certain types of food since interactions may occur. Using alcohol or tobacco with certain medicines may also cause interactions to occur. Discuss with your healthcare professional the use of your medicine with food, alcohol, or tobacco.

L-glutamine common side effects.

Tell your doctor if any of these symptoms are severe or do not go away:

  • constipation
  • nausea
  • headache
  • abdominal pain
  • cough
  • back, leg, feet, hands, or arm pain

L-glutamine may cause other side effects. See your doctor if you have any unusual problems while taking this medication.

Less common side effects

  • Blood in urine
  • changes in skin color
  • chills
  • cold hands and feet
  • confusion
  • cough
  • difficulty swallowing
  • dizziness
  • fainting
  • fast heartbeat
  • fever
  • frequent and painful urination
  • headache
  • hives, itching, skin rash
  • light headedness
  • lower back or side pain
  • pain, redness, or swelling in the arm or leg
  • puffiness or swelling of the eyelids or around the eyes, face, lips, or tongue
  • rapid, shallow breathing
  • stomach pain
  • sudden decrease in amount of urine
  • tightness in the chest
  • unusual tiredness or weakness
  • wheezing

Some side effects may occur that usually do not need medical attention. These side effects may go away during treatment as your body adjusts to the medicine. Also, your health care professional may be able to tell you about ways to prevent or reduce some of these side effects. Check with your health care professional if any of the following side effects continue or are bothersome or if you have any questions about them.

Other Less common side effects

    1. Abnormal or decreased touch sensation
    2. back pain
    3. bacterial infection
    4. bleeding after defecation
    5. bleeding, blistering, burning, coldness, discoloration of skin, feeling of pressure, hives, infection, inflammation, itching, lumps, numbness, pain, rash, redness, scarring, soreness, stinging, swelling, tenderness, tingling, ulceration, or warmth at site
    6. bloated full feeling
    7. body aches or pain
    8. breast pain, female
    9. chest pain
    10. change in the color, amount, or odor of vaginal discharge
    11. congestion
    12. constipation
    13. Crohn’s disease, aggravated
    14. dark urine
    15. decreased urination
    16. diarrhea
    17. difficulty having a bowel movement (stool)
    18. difficulty in moving
    19. discoloration of fingernails or toenails
    20. discouragement
    21. dry mouth
    22. dryness or soreness of throat
    23. ear or hearing symptoms
    24. excess air or gas in stomach or intestines
    25. feeling sad or empty
    26. feeling unusually cold shivering
    27. flatulence
    28. full or bloated feeling
    29. general feeling of discomfort or illness
    30. increase in heart rate
    31. indigestion
    32. irritability
    33. joint pain;
    34. lack of appetite
    35. light-colored stools
    36. loss of appetite
    37. loss of interest or pleasure
    38. muscle aches and pains
    39. muscle pain or stiffness
    40. nausea
    41. pain in joints
    42. pain or burning while urinating
    43. pains in stomach, side, or abdomen, possibly radiating to the back
    44. passing gas
    45. pressure in the stomach
    46. rash
    47. rectal bleeding
    48. runny nose
    49. shivering
    50. sleeplessness
    51. sneezing
    52. sore throat
    53. stomach bloating, burning, cramping, or pain
    54. stuffy nose
    55. sunken eyes
    56. sweating
    57. swelling of abdominal or stomach area
    58. swelling of face
    59. swelling of hands, ankles, feet, or lower legs
    60. swollen joints
    61. tender, swollen glands in neck;
    62. thirst
    63. trouble concentrating
    64. trouble sleeping
    65. trouble in swallowing
    66. unable to sleep
    67. uncomfortable swelling around anus
    68. unpleasant breath odor
    69. unusual tiredness or weakness
    70. voice changes
    71. vomiting
    72. vomiting of blood
    73. weight loss
    74. wrinkled skin
    75. yellow eyes or skin

Other side effects not listed may also occur in some patients. If you notice any other effects, check with your healthcare professional.

Call your doctor for medical advice about side effects.

References
  1. L-glutamine. https://pubchem.ncbi.nlm.nih.gov/compound/L-glutamine
  2. Gaurav K, Goel RK, Shukla M, Pandey M. Glutamine: A novel approach to chemotherapy-induced toxicity. Indian Journal of Medical and Paediatric Oncology : Official Journal of Indian Society of Medical & Paediatric Oncology. 2012;33(1):13-20. doi:10.4103/0971-5851.96962. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3385273/
  3. Intracellular free amino acid concentration in human muscle tissue. Bergström J, Fürst P, Norée LO, Vinnars E. J Appl Physiol. 1974 Jun; 36(6):693-7. https://www.ncbi.nlm.nih.gov/pubmed/4829908/
  4. Is glutamine a conditionally essential amino acid? Lacey JM, Wilmore DW. Nutr Rev. 1990 Aug; 48(8):297-309. https://www.ncbi.nlm.nih.gov/pubmed/2080048/
  5. L-glutamine. Medline Plus. https://medlineplus.gov/druginfo/meds/a617035.html
  6. Souba WW. Glutamine Physiology, Biochemistry, and Nutrition in Critical Illness. Austin, TX: R.G. Landes Co; 1992.
  7. Halperin HL, Kamel HS, Ethier JH. Biochemistry and physiology of ammonium excretion. In: Seldin DW, Giebisch L, editors. The kidney: Physiology and pathology. New York: Raven Press; 1992. pp. 2645–80
  8. Polyglutamine aggregates alter protein folding homeostasis in Caenorhabditis elegans. Satyal SH, Schmidt E, Kitagawa K, Sondheimer N, Lindquist S, Kramer JM, Morimoto RI. Proc Natl Acad Sci U S A. 2000 May 23; 97(11):5750-5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC18505/
  9. Bacterial and yeast chaperones reduce both aggregate formation and cell death in mammalian cell models of Huntington’s disease. Carmichael J, Chatellier J, Woolfson A, Milstein C, Fersht AR, Rubinsztein DC. Proc Natl Acad Sci U S A. 2000 Aug 15; 97(17):9701-5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC16928/
  10. Blomqvist BI, Hammarqvist F, von der Decken A, Wernerman J. Glutamine and alpha-ketoglutarate prevent the decrease in muscle free glutamine concentration and influence protein synthesis after total hip replacement. Metabolism. 1995;44:1215–22. https://www.ncbi.nlm.nih.gov/pubmed/7666798
  11. Planas M, Schwartz S, Arbós MA, Farriol M. Plasma glutamine levels in septic patients. J Parenter Enteral Nutr. 1993;17:299–300. https://www.ncbi.nlm.nih.gov/pubmed/8505843
  12. Wernerman J, Hammarkvist F, Ali MR, Vinnars E. Glutamine and ornithine-alpha-ketoglutarate but not branched-chain amino acids reduce the loss of muscle glutamine after surgical trauma. Metabolism. 1989;38:63–6. https://www.ncbi.nlm.nih.gov/pubmed/2503684
  13. Parry-Billings M, Evans J, Calder PC, Newsholme EA. Does glutamine contribute to immunosuppression after major burns? Lancet. 1990;336:523–5. https://www.ncbi.nlm.nih.gov/pubmed/1975037
  14. Ziegler TR. Glutamine and Acute Illness. Br J Nutr. 2002;87(Suppl 1):S9–15. https://www.ncbi.nlm.nih.gov/pubmed/11895159
  15. Thompson JS, Rochling FA, Weseman RA, Mercer DF. Current management of short bowel syndrome. Current Problems in Surgery. 2012;49(2):52–115.
  16. Goringe AP, Brown S, O’Callaghan U, Rees J, Jebb S, Elia M, et al. Glutamine and vitamin E in the treatment of hepatic veno-occlusive disease following high-dose chemotherapy. Bone Marrow Transplant. 1998;22:2879–84. https://www.ncbi.nlm.nih.gov/pubmed/9603409
  17. Muscaritoli M, Grieco G, Capria S, Iori AP, Rossi Fanelli F. Nutritional and metabolic support in patients undergoing bone marrow transplantation. Am J Clin Nutr. 2002;75:183–90. http://ajcn.nutrition.org/content/75/2/183.long
  18. Miller AL. Therapeutic considerations of L-glutamine: A review of the literature. Alternat Med Rev. 1999;4:239–48. https://www.ncbi.nlm.nih.gov/pubmed/10468648
  19. Kang YJ, Feng Y, Hatcher EL. Glutathione stimulates A549 cell proliferation in glutamine-deficient culture: The effect of glutamine supplementation. J Cell Physiol. 1994;161:589–96. https://www.ncbi.nlm.nih.gov/pubmed/7962140
  20. Bartlett DL, Charland S, Torosian MH. Effect of glutamine on tumor and host growth. Ann Surg Oncol. 1995;2:71–6. https://www.ncbi.nlm.nih.gov/pubmed/7530589
  21. Van der Hulst RR, von Meyenfeldt MF, Deutz NE, Soeters PB. Glutamine extraction by the gut is reduced in depleted [corrected] patients with gastrointestinal cancer. Annals of Surgery. 1997;225(1):112-121. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1190613/
  22. Klimberg VS, Souba WW, Dolson DJ, Salloum RM, Hautamaki RD, Plumley DA, et al. Prophylactic glutamine protects the intestinal mucosa from radiation injury. Cancer. 1990;66:62–8. https://www.ncbi.nlm.nih.gov/pubmed/2354410
  23. Fox AD, Kripke SA, De Paula J, Berman JM, Settle RG, Rombeau JL. Effect of a glutamine-supplemented enteral diet on methotrexate-induced enterocolitis. JPEN J Parenter Enteral Nutr. 1988;12:325–31. https://www.ncbi.nlm.nih.gov/pubmed/3138440
  24. Klimberg VS, Souba WW, Salloum RM, Plumley DA, Cohen FS, Dolson DJ, et al. Glutamine-enriched diets support muscle glutamine metabolism without stimulating tumor growth. J Surg Res. 1990;48:319–23. https://www.ncbi.nlm.nih.gov/pubmed/2338817
  25. Smith CV, Mitchell JR. Pharmacological aspects of glutathione in drug metabolism. In: Dolphin D, Poulson R, Avramovic O, editors. Coenzymes and Cofactors. New York: John Wiley and Sons; 1989. pp. 1–44.
  26. Nirenberg A, Mosende C, Mehta BM, Gisolfi AL, Rosen G. High-dose methotrexate concentrations and corrective measures to avert toxicity. Cancer Treat Rep. 1977;61:779–83. https://www.ncbi.nlm.nih.gov/pubmed/302143
  27. Jensen JC, Nwokedi E, Baker ML, Bevans DW, 3rd, Baker ML, Pappas AA, et al. Prevention of chronic radiation enteropathy by dietary glutamine. Ann Surg Oncol. 1994;1:157–63. https://www.ncbi.nlm.nih.gov/pubmed/7834441
  28. Rouse K, Nwokedi E, Woodliff JE, Epstein J, Klimberg VS. Glutamine enhances selectivity of chemotherapy through changes in glutathione metabolism. Annals of Surgery. 1995;221(4):420-426. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1234593/
  29. Scheid C, Hermann K, Kremer G, Holsing A, Heck G, Fuchs M, et al. Randomized, double-blind, controlled study of glycyl-glutamine-dipeptide in the parenteral nutrition of patients with acute leukemia undergoing intensive chemotherapy. Nutrition. 2004;20:249–54. https://www.ncbi.nlm.nih.gov/pubmed/14990264
  30. GLUTAMINE. https://www.webmd.com/vitamins-supplements/ingredientmono-878-glutamine.aspx?activeingredientid=878
  31. Glutamine (Oral route). https://www.ncbi.nlm.nih.gov/pubmedhealth/PMH0046028/
read more
12 Body SystemsImmune and Hematology System

What are white blood cells

white blood cells

What are white blood cells

White blood cells, also called leukocytes (or leucocytes), are the cells of the immune system that are involved in protecting the body against both infectious disease and foreign invaders 1. All white blood cells are produced and derived from multipotent cells in the bone marrow known as hematopoietic stem cells. Leukocytes are found throughout the body, including the blood and lymphatic system.

Most blood samples are about 45% red blood cells by volume, with the white blood cells and platelets accounting for less than 1% of blood volume. The remaining blood sample, about 55%, is the plasma, a clear, straw-colored liquid (see Figure 1). Blood transports leukocytes to sites of infection. Leukocytes may then leave the bloodstream.

Like red blood cells, white blood cells (leukocytes) have limited life spans, so they must constantly be replaced. Leukocytes develop from hematopoietic stem cells in the red bone marrow (see Figure 2) in response to hormones, much as red blood cells form from precursors upon stimulation from erythropoietin. The hormones that affect leukocyte development fall into two groups—interleukins and colony-stimulating factors (CSFs). Interleukins are numbered, while most colony stimulating factors are named for the cell population they stimulate.

Types of white blood cells

All white blood cells have nuclei, which distinguishes them from the other blood cells, the anucleated red blood cells and platelets. They differ in size, the nature of their cytoplasm, the shape of the nucleus, and their staining characteristics, and they are named for these distinctions. Two pairs of broadest categories classify them either by structure (granulocytes or agranulocytes) or by cell division lineage (myeloid cells or lymphoid cells) (see Figure 3). These broadest categories can be further divided into the five main types: neutrophils, eosinophils, basophils, lymphocytes, and monocytes 2. These types are distinguished by their physical and functional characteristics. Monocytes and neutrophils are phagocytic. Further subtypes can be classified; for example, among lymphocytes, there are B cells, T cells, and NK (natural killer) cells.

Normally, five types of white blood cells are in circulating blood. For example, leukocytes with granular cytoplasm are called granulocytes, whereas those without cytoplasmic granules are called agranulocytes (see Figure. 3).

A typical granulocyte is about twice the size of a red blood cell. Members of this group include neutrophils, eosinophils, and basophils. Granulocytes develop in red bone marrow as do red blood cells, but they have short life spans, averaging about 12 hours.

Neutrophils have fine cytoplasmic granules that appear light purple in neutral stain. The nucleus of an older neutrophil is lobed and consists of two to five sections (segments, so these cells are sometimes called segs) connected by thin strands of chromatin (Figure 4). Younger neutrophils are also called bands because their nuclei are C-shaped. Neutrophils account for 54% to 62% of the leukocytes in a typical blood sample from an adult.

Eosinophils contain coarse, uniformly sized cytoplasmic granules that appear deep red in acid stain (Figure 4). The nucleus usually has only two lobes (termed
bilobed). Eosinophils make up 1% to 3% of the total number of circulating leukocytes.

Basophils are similar to eosinophils in size and in the shape of their nuclei, but they have fewer, more irregularly shaped cytoplasmic granules that appear deep
blue in basic stain (Figure 4). Basophils usually account for less than 1% of the circulating leukocytes.

The leukocytes of the agranulocyte group include monocytes and lymphocytes. Monocytes generally arise from red bone marrow. Lymphocytes are formed in the organs of the lymphatic system, as well as in the red bone marrow.

Monocytes, the largest blood cells, are two to three times greater in diameter than red blood cells. Their nuclei are round, kidney-shaped, oval, or lobed. They usually make up 3% to 9% of the leukocytes in a blood sample and live for several weeks or even months.

Lymphocytes are only slightly larger than red blood cells. A typical lymphocyte has a large, round nucleus surrounded by a thin rim of cytoplasm. These cells account for 25% to 33% of circulating leukocytes. Lymphocytes may live for years.

Figure 1. Blood composition

Note: Blood is a complex mixture of formed elements in a liquid extracellular matrix, called blood plasma. Note that water and proteins account for 99% of the blood plasma.

blood composition

blood compositionblood-composition

Note: Blood consists of a liquid portion called plasma and a solid portion (the formed elements) that includes red blood cells, white blood cells, and platelets. When blood components are separated by centrifugation, the white blood cells and platelets form a thin layer, called the “buffy coat,” between the plasma and the red blood cells, which accounts for about 1% of the total blood volume. Blood cells and platelets can be seen under a light microscope when a blood sample is smeared onto a glass slide.

Normal Leukocyte Life Cycle and Responses

The life cycle of leukocytes includes development and differentiation, storage in the bone marrow, margination within the vascular spaces, and migration to tissues. Stem cells in the bone marrow produce cell lines of erythroblasts, which become red blood cells; megakaryoblasts, which become platelets; lymphoblasts; and myeloblasts. Lymphoblasts develop into various types of T and B cell lymphocytes. Myeloblasts further differentiate into monocytes and granulocytes, a designation that includes neutrophils, basophils, and eosinophils (Figure 2 and 3). Once white blood cells have matured within the bone marrow, 80% to 90% remain in storage in the bone marrow. This large reserve allows for a rapid increase in the circulating white blood cell count within hours. A relatively small pool (2% to 3%) of leukocytes circulate freely in the peripheral blood 3; the rest stay deposited along the margins of blood vessel walls or in the spleen. Leukocytes spend most of their life span in storage. Once a leukocyte is released into circulation and peripheral tissues, its life span ranges from two to 16 days, depending on the type of cell.

Figure 2. Bone marrow anatomy

bone marrow anatomy

Anatomy of the bone. The bone is made up of compact bone, spongy bone, and bone marrow. Compact bone makes up the outer layer of the bone. Spongy bone is found mostly at the ends of bones and contains red marrow. Bone marrow is found in the center of most bones and has many blood vessels. There are two types of bone marrow: red and yellow. Red marrow contains blood stem cells that can become red blood cells, white blood cells, or platelets. Yellow marrow is made mostly of fat.

Figure 3. White blood cells development. A blood stem cell goes through several steps to become a red blood cell, platelet, or white blood cell

blood cell development

Figure 4. White blood cells development

white blood cells development

Figure 5. White blood cells

white blood cells

White Blood Cell Counts

The number of leukocytes in a microliter of human blood, called the white blood cell count (WBCC or WCC), normally is 3,500 to 10,500 per mm3 (3.5 to 10.5 × 109 per L). White blood cell counts are of clinical interest because their number may change in response to abnormal conditions. A total number of white blood cells exceeding 10,500 per mm3 (10.5 × 109 per L) of blood constitutes leukocytosis, indicating acute infection, such as appendicitis. The white blood cell count is greatly elevated in leukemia.

A total white blood cell count below 3,500 per mm3 (<3.5 × 109 per L) of blood is called leukopenia. Such a deficiency may accompany typhoid fever, influenza, measles, mumps, chickenpox, AIDS, or poliomyelitis.

Healthy newborn infants may have a white blood cell count from 13,000 to 38,000 per mm3 (13.0 to 38.0 × 109 per L) at 12 hours of life. By two weeks of age, this decreases to approximately 5,000 to 20,000 per mm3 (5.0 to 20.0 × 109 per L), and gradually declines throughout childhood to reach adult levels of 4,500 to 11,000 per mm3 (4.5 to 11.0 × 109 per L) by about 21 years of age 4. There is also a shift from relative lymphocyte to neutrophil predominance from early childhood to the teenage years and adulthood 5. During pregnancy, there is a gradual increase in the normal white blood cell count (third trimester 95% upper limit = 13,200 per mm3 [13.2 × 109 per L] and 99% upper limit = 15,900 per mm3 [15.9 × 109 per L]), and a slight shift toward an increased percentage of neutrophils 6. In one study of afebrile postpartum patients, the mean white blood cell count was 12,620 per mm3 (12.62 × 109 per L) for women after vaginal deliveries and 12,710 per mm3 (12.71 × 109 per L) after cesarean deliveries. Of note, positive bacterial cultures were not associated with leukocytosis or neutrophilia, making leukocytosis an unreliable discriminator in deciding which postpartum patients require antibiotic therapy 7. Patients of black African descent tend to have a lower white blood cell count (by 1,000 per mm3 [1.0 × 109 per L]) and lower absolute neutrophil counts 8.

A differential white blood cell count lists percentages of the various types of leukocytes in a blood sample. This test is useful because the relative proportions of white blood cells may change in particular diseases. The number of neutrophils, for instance, usually increases during bacterial infections. Eosinophils may become more abundant during certain parasitic infections and allergic reactions. In AIDS, the number of a type of lymphocyte called T cells drops sharply.

Table 1. White Blood Cell Count Variation with Age and Pregnancy

Patient characteristicNormal total leukocyte count

Newborn infant

13,000 to 38,000 per mm3 (13.0 to 38.0 × 109 per L)

Infant two weeks of age

5,000 to 20,000 per mm3 (5.0 to 20.0 × 109 per L)

Adult

4,500 to 11,000 per mm3 (4.5 to 11.0 × 109 per L)

Pregnant female (third trimester)

5,800 to 13,200 per mm3 (5.8 to 13.2 × 109 per L)

[Source 4]

Differentiation by Type of White Blood Cell

Changes in the normal distribution of types of white blood cells can indicate specific causes of leukocytosis 9. Although the differential of the major types of white blood cells is important for evaluating the cause of leukocytosis, it is sometimes helpful to think in terms of absolute, rather than relative, leukopenias and leukocytoses. To calculate the absolute cell count, the total leukocyte count is multiplied by the differential percentage. For example, with a normal white blood cell count of 10,000 per mm3 (10.0 × 109 per L) and an elevated monocyte percentage of 12, the absolute monocyte count is 12% or 0.12 times the white blood cell count of 10,000 per mm3, yielding 1,200 per mm3 (1.2 × 109 per L), which is abnormally elevated.

Table 2. Normal White Blood Cell Distribution

White blood cell lineNormal percentage of total leukocyte count

Neutrophils

40 to 60

Lymphocytes

20 to 40

Monocytes

2 to 8

Eosinophils

1 to 4

Basophils

0.5 to 1

[Source 9]

Functions of White Blood Cells

White blood cells protect your body against infection in various ways. Some leukocytes phagocytize (engulfing and destroying bacteria and other foreign material) bacterial cells in the body, and others produce proteins (antibodies) that destroy or disable foreign particles.

Leukocytes can squeeze between the cells that form the walls of the smallest blood vessels. This movement, called diapedesis, allows the white blood cells to leave the circulation. Once outside the blood, they move through interstitial spaces using a form of self propulsion called amoeboid motion.

The most mobile and active phagocytic leukocytes are neutrophils and monocytes. Monocytes leave the bloodstream and specialize further to become macrophages that phagocytize bacteria, dead cells, and other debris in the tissues.

Neutrophils cannot ingest particles much larger than bacterial cells, but monocytes can engulf large objects. Both of these phagocytes contain many lysosomes, which are organelles filled with digestive enzymes that break down organic molecules in captured bacteria, nutrients, and wornout organelles. Neutrophils and monocytes may become so engorged with digestive products and bacterial toxins that they die.

Eosinophils are only weakly phagocytic, but they are attracted to and can kill certain parasites. Eosinophils also help control inflammation and allergic reactions by removing biochemicals associated with these reactions.

Basophils migrate to damaged tissues, where they release heparin, which inhibits blood clotting, and histamine, which promotes inflammation. Basophils also play major roles in certain allergic reactions.

Lymphocytes are important in immunity. Lymphocytes called B cells, for example, produce antibodies that attack specific foreign substances that enter the body.

Phagocytosis

Phagocytosis removes foreign particles from the lymph as it moves from the interstitial spaces to the bloodstream. Phagocytes in the blood vessels and in the tissues of the spleen, liver, or bone marrow remove particles that reach the blood. The blood’s most active phagocytic cells are neutrophils and monocytes. Chemicals released from injured tissues attract these cells by chemotaxis. Neutrophils engulf and digest smaller particles; monocytes phagocytize larger ones.

Monocytes that leave the bloodstream by diapedesis become macrophages. These large cells may be free, or fixed in various tissues. The fixed macrophages can divide and produce new macrophages. Neutrophils, monocytes and macrophages constitute the mononuclear phagocytic system (reticuloendothelial system).

Table 3. White blood cells types and functions

TypeApprox. % in adultsDiameter (μm)Main targetsNucleusGranulesLifetime
Neutrophil62%10–12
  • Bacteria
  • Fungi
MultilobedFine, faintly pink (H&E stain)6 hours–few days
(days in spleen and other tissue)
Eosinophil2.3%10–12
  • Larger parasites
  • Modulate allergic inflammatory responses
Bi-lobedFull of pink-orange (H&E stain)8–12 days (circulate for 4–5 hours)
Basophil0.4%12–15
  • Release histamine for inflammatory responses
Bi-lobed or tri-lobedLarge blueA few hours to a few days
Lymphocyte30%Small lymphocytes 7–8 Large lymphocytes 12–15
  • B cells: releases antibodies and assists activation of T cells
  • T cells:
    • CD4+ Th (T helper) cells: activate and regulate T and B cells
    • CD8+ cytotoxic T cells: virus-infected and tumor cells.
    • γδ T cells: bridge between innate and adaptive immune responses; phagocytosis
    • Regulatory (suppressor) T cells: Returns the functioning of the immune system to normal operation after infection; prevents autoimmunity
  • Natural killer cells: virus-infected and tumor cells.
Deeply staining, eccentricNK-cells and cytotoxic (CD8+) T-cellsYears for memory cells, weeks for all else.
Monocyte5.3%15–30Monocytes migrate from the bloodstream to other tissues and differentiate into tissue resident macrophages, Kupffer cells in the liver.Kidney shapedNoneHours to days
[Source 10]

Lymphatic tissue contains lymphocytes, macrophages, and other cells.

The unencapsulated diffuse lymphatic tissue associated with the digestive, respiratory, urinary, and reproductive tracts is called the mucosa-associated lymphoid tissue (MALT). Compact masses of lymphatic tissue compose the tonsils and appendix. Mucosa-associated lymphoid tissue (MALT) aggregates of lymphatic tissue, called Peyer’s patches, are scattered throughout the mucosal lining of the distal portion of the small intestine. The lymphatic organs, including the lymph nodes, thymus, and spleen, are encapsulated lymphatic tissue. A capsule of connective tissue with many fibers encloses each organ.

Lymph Nodes

Lymph nodes contain large numbers of lymphocytes (B cells and T cells) and macrophages that fight invading microorganisms. Masses of B cells and macrophages in the cortex are contained within lymphatic nodules, also called lymphatic follicles, the functional units of the lymph node. The spaces within a node, called lymphatic sinuses, provide a complex network of chambers and channels through which lymph circulates.

Lymph nodes are generally in groups or chains along the paths of the larger lymphatic vessels throughout the body, but are absent in the central nervous system. Figure 5 shows the major locations of lymph nodes.

Lymph nodes have two primary functions: (1) filtering potentially harmful particles from lymph before returning it to the bloodstream, and (2) monitoring body fluids (immune surveillance), a function performed by lymphocytes and macrophages. Along with red bone marrow, the lymph nodes are centers for lymphocyte production. Lymphocytes attack viruses, bacteria, and other parasitic cells that are brought to the lymph nodes by lymph in the lymphatic vessels. Macrophages in the lymph nodes engulf and destroy foreign substances, damaged cells, and cellular debris.

Superficial lymphatic vessels inflamed by bacterial infection appear as red streaks beneath the skin, a condition called lymphangitis. Inflammation of the lymph nodes, called lymphadenitis, often follows. In lymphadenopathy, affected lymph nodes enlarge and may be quite painful.

Figure 5. Locations of major lymph nodes

Locations of major lymph nodes

Thymus

The thymus is a soft, bilobed gland enclosed in a connective tissue capsule and located anterior to the aorta. It is posterior to the upper part of the sternum. The
thymus is usually proportionately larger during infancy and early childhood, but shrinks after puberty and may be quite small in an adult. In elderly people, adipose and connective tissues replace lymphatic tissue in the thymus. In a person aged seventy, the thymus is one-tenth the size it was in that person at the age of ten.

Connective tissues extend inward from the surface of the thymus, subdividing it into lobules. The lobules house many lymphocytes. Most of these cells (thymocytes) are inactive; however, some mature into T lymphocytes (T cells), which leave the thymus and provide immunity. Epithelial cells in the thymus secrete hormones called thymosins, which stimulate maturation of T lymphocytes.

Figure 6. Thymus

thymus

Spleen

The spleen, the largest lymphatic organ, is in the upper left portion of the abdominal cavity, just inferior to the diaphragm. It is posterior and lateral to the stomach. The spleen resembles a large lymph node and is subdivided into lobules. However, unlike the lymphatic sinuses of a lymph node, the spaces in the spleen, called venous sinuses, are filled with blood instead of lymph.

The tissues within splenic lobules are of two types (Figure 7). The white pulp is distributed throughout the spleen in tiny islands. This tissue is composed of splenic nodules, which are similar to the lymphatic nodules in lymph nodes and are packed with lymphocytes. The red pulp, which fills the remaining spaces of the lobules, surrounds the venous sinuses. This pulp contains numerous red blood cells, which impart its color, plus many lymphocytes and macrophages.

Blood capillaries in the red pulp are quite permeable. Red blood cells can squeeze through the pores in these capillary walls and enter the venous sinuses. The older, more fragile red blood cells may rupture during this passage, and the resulting cellular debris is removed by phagocytic macrophages in the venous sinuses. These macrophages also engulf and destroy foreign particles, such as bacteria, that may be carried in the blood as it flows through the venous sinuses. Thus, the spleen filters blood much as the lymph nodes filter lymph.

Figure 7. The Spleen

spleen

Body Defenses Against Infection

The presence and multiplication of a disease-causing agent, or pathogen, which if unchecked may cause an infection. Pathogens include viruses, bacteria, fungi, and protozoans.

The human body can prevent the entry of pathogens or destroy them if they enter. Some mechanisms are general in that they protect against many types of pathogens, providing innate (nonspecific) defense. These mechanisms include species resistance, mechanical barriers, inflammation, chemical barriers (enzyme action, interferon, and complement), natural killer cells, phagocytosis, and fever.

Other defense mechanisms are very precise, targeting specific pathogens and providing adaptive (specific) defense, or immunity. Specialized  lymphocytes that recognize foreign molecules (nonself antigens) in the body act against the foreign molecules in several ways, including the production of cytokines and antibodies. Innate and adaptive defense mechanisms work together to protect the body against infection. The innate defenses respond quite  rapidly, while adaptive defenses develop more slowly.

Inflammation

Inflammation is a tissue response to injury or infection, producing localized redness, swelling, heat, and pain. The redness is a result of blood vessel dilation that increases blood flow and volume in the affected tissues. This effect, coupled with an increase in the permeability of nearby capillaries and subsequent leakage of protein-rich fluid into tissue spaces, swells tissues (edema). The heat comes as blood enters from deeper body parts, which are warmer than the surface. Pain results from stimulation of nearby pain receptors.

Infected cells release chemicals that attract white blood cells to sites of inflammation. Here the white blood cells phagocytize pathogens. Local heat speeds up phagocytic activity. In bacterial infections, the resulting mass of white blood cells, bacterial cells, and damaged tissue may form a thick fluid called pus.

Fluids that leak out of the capillaries, called exudates, also collect in inflamed tissues. These fluids contain fibrinogen and other blood-clotting factors. Clotting forms a network of fibrin threads in the affected region. Later, fibroblasts may arrive and secrete matrix components until the area is enclosed in a connective  tissue sac. This walling off of the infected area helps inhibit the spread of pathogens and toxins to adjacent tissues.

Chemical Barriers

Lymphocytes and fibroblasts produce hormone like peptides called interferons in response to viruses or tumor cells. Once released from the virus-infected cell, interferon binds to receptors on uninfected cells, stimulating them to synthesize proteins that block replication of a variety of viruses. Thus, interferon’s effect is nonspecific. Interferons also stimulate phagocytosis and enhance the activity of other cells that help resist infections and the growth of tumors.

Complement is a group of proteins, in plasma and other body fluids, that interact in an expanding series of reactions or cascade. Activation of complement stimulates inflammation, attracts phagocytes, and enhances phagocytosis.

Natural Killer (NK) Cells

Natural killer (NK) cells are a small population of lymphocytes. They are different from the lymphocytes (T and B cells) that provide adaptive (specific) defense mechanisms. NK cells defend the body against various viruses and cancer cells by secreting cytolytic (“cell-cutting”) substances called perforins that lyse the cell membrane, destroying the infected cell. NK cells also secrete chemicals that enhance inflammation.

Fever

Fever is body temperature elevated above an individual’s normal temperature due to an elevated setpoint. It is part of the innate defense because as a result of the fever the body becomes inhospitable to certain pathogens. Higher body temperature causes the liver and spleen to sequester iron, which reduces the level of iron in the blood. Because bacteria and fungi require iron for normal metabolism, their growth and reproduction in a fever-ridden body slows and may cease. Also, phagocytic cells attack more vigorously when the temperature rises. For these reasons, low-grade fever of short duration may be a natural response to infection, not a treated symptom.

Immunity

The third line of defense, immunity, is resistance to specific pathogens or to their toxins or metabolic by-products. Lymphocytes and macrophages that recognize and remember specific foreign molecules carry out adaptive immune responses, which include the cellular immune response and the humoral immune response.

Antigens

Antigens are proteins, polysaccharides, glycoproteins, or glycolipids that can elicit an immune response. Before birth, cells inventory the antigens in the body, learning to identify these as “self.” The immune response is to “nonself,” or foreign, antigens, but not normally to self antigens. Receptors on lymphocyte surfaces enable these cells to recognize nonself antigens.

The antigens most effective in eliciting an immune response are large and complex, with few repeating parts. A smaller molecule that cannot by itself stimulate an immune response may combine with a larger molecule, which makes it detectable. Such a small molecule is called a hapten. Stimulated lymphocytes react either to the hapten or to the larger molecule of the combination. Hapten molecules are in drugs such as penicillin, in household and industrial chemicals, in dust particles, and in animal dander.

Lymphocyte Origins

Lymphocyte production begins during fetal development and continues throughout life, with red bone marrow releasing unspecialized precursors to lymphocytes into the circulation. About half of these cells reach the thymus, where they specialize into T lymphocytes, or T cells. After leaving the thymus, some of these T cells constitute 70% to 80% of the circulating lymphocytes in blood. Other T cells reside in lymphatic organs and are particularly abundant in the lymph nodes, thoracic duct, and white pulp of the spleen.

Other lymphocytes remain in the red bone marrow until they differentiate into B lymphocytes, or B cells. The blood distributes B cells, which constitute 20% to 30% of circulating lymphocytes. B cells settle in lymphatic organs along with T cells and are abundant in the lymph nodes, spleen, bone marrow, and intestinal lining. Figure 8 illustrates B cell and T cell production. Table 1 compares the characteristics of T cells and B cells.

Figure 8. Lymphocyte production

lymphocyte production

T Cells and the Cellular Immune Response

A lymphocyte must be activated before it can respond to an antigen. T cell activation requires that processed fragments of the antigen be attached to the surface of another type of cell, called an antigen-presenting cell (accessory cell). Macrophages, B cells, and several other cell types can be antigen-presenting cells.

T cell activation may occur when a macrophage phagocytizes a bacterium and digests it within a phagolysosome formed by the fusion of the vesicle containing the bacterium (phagosome) and a lysosome. Some of the resulting bacterial antigens are then displayed on the macrophage’s cell membrane near certain protein molecules that are part of a group of proteins called the major histocompatibility complex (MHC). MHC antigens help T cells recognize that a newly displayed antigen is foreign (nonself).

Activated T cells interact directly with antigen-bearing cells. Such cell-to-cell contact is called the cellular immune response, or cell-mediated immunity. T cells (and some macrophages) also synthesize and secrete polypeptides called cytokines that enhance certain cellular responses to antigens. For example, interleukin-1 and interleukin-2 stimulate the synthesis of several other cytokines from other T cells. Additionally, interleukin-1 helps activate T cells, whereas interleukin-2 causes T cells to proliferate. This proliferation increases the number of T cells in a clone, which is a group of genetically identical cells that descend from a single, original cell. Other cytokines, called colony stimulating factors (CSFs), stimulate leukocyte production in red bone marrow and activate macrophages. T cells may also secrete toxins that kill their antigen-bearing target cells, growth-inhibiting factors that prevent target cell growth, or interferon that inhibits the proliferation of viruses and tumor cells. Several types of T cells have distinct functions.

A specialized type of T cell, called a helper T cell, is activated when its antigen receptor combines with a displayed foreign antigen (Figure 9). Once activated, the
helper T cell proliferates and the resulting cells stimulate B cells to produce antibodies that are specific for the displayed antigen.

Another type of T cell is a cytotoxic T cell, which recognizes and combines with nonself antigens that cancerous cells or virally infected cells display on their surfaces near certain MHC proteins. Cytokines from helper T cells activate the cytotoxic T cell. Next, the cytotoxic T cell proliferates. Cytotoxic T cells then bind to the surfaces of antigen-bearing cells, where they release perforin protein that cuts pore like openings in the cell membrane, destroying these cells. In this way, cytotoxic T cells continually monitor the body’s cells, recognizing and eliminating tumor cells and cells infected with viruses. Cytotoxic T cells provide much of the body’s defense against HIV infection.

Some cytotoxic T cells do not respond to a nonself antigen on first exposure, but remain as memory T cells that provide for future immune protection. Upon subsequent exposure to the same antigen, these memory cells immediately divide to yield more cytotoxic T cells and helper T cells, often before symptoms arise.

Figure 9. T-cell activation

t-cell activation

B Cells and the Humoral Immune Response

When a B cell encounters an antigen whose molecular shape fits the shape of the B cell’s antigen receptors, it becomes activated. In response to the receptor-antigen combination, the B cell divides repeatedly, expanding its clone. However, most of the time B cell activation requires T cell “help.” When an activated helper T cell encounters a B cell already combined with a foreign antigen identical to the one that activated the helper T cell, the helper T cell releases certain cytokines. These cytokines stimulate the B cell to proliferate, increasing the number of cells in its clone of antibody-producing cells (Figure 10). The cytokines also attract macrophages and leukocytes into inflamed tissues and help keep them there.

Some members of the activated B cell’s clone differentiate further into plasma cells, which produce and secrete large globular proteins called antibodies, also called immunoglobulins. Antibodies are similar in structure to the antigen-receptor molecules on the original B cell’s surface. Body fluids carry antibodies, which then react in various ways to destroy specific antigens or antigen-bearing particles. This antibody-mediated immune response is called the humoral immune response.

An individual’s B cells can produce more than 1,000,000,000 different antibodies, each reacting against a specific antigen. The enormity and diversity of the antibody response defends against many pathogens.

Other members of the activated B cell’s clone differentiate further into memory B cells. Like memory T cells, these memory B cells respond rapidly to subsequent exposure to a specific antigen.

Table 2 summarizes the steps leading to antibody production as a result of B cell and T cell activities.

Table 2. Steps in Antibody Production

B Cell Activities
  1. Antigen-bearing agents enter tissues.
  2. B cell encounters an antigen that fits its antigen receptors.
  3. Either alone or more often in conjunction with helper T cells, the B cell is activated. The B cell proliferates, enlarging its clone.
  4. Some of the newly formed B cells differentiate further to become plasma cells.
  5. Plasma cells synthesize and secrete antibodies whose molecular structure is similar to the activated B cell’s antigen receptors.
T Cell Activities
  1. Antigen-bearing agents enter tissues.
  2. An accessory cell, such as a macrophage, phagocytizes the antigen-bearing agent, and the macrophage’s lysosomes digest the agent.
  3. Antigens from the digested antigen-bearing agents are displayed on the membrane of the accessory cell.
  4. Helper T cell becomes activated when it encounters a displayed antigen that fits its antigen receptors.
  5. Activated helper T cell releases cytokines when it encounters a B cell that has previously combined with an identical antigen-bearing agent.
  6. Cytokines stimulate the B cell to proliferate, enlarging its clone.
  7. Some of the newly formed B cells give rise to cells that differentiate into antibody-secreting plasma cells.

Figure 10. B-cell activation

b-cell activation

Types of Antibodies

Antibodies (immunoglobulins) are soluble, globular proteins that constitute the gamma globulin fraction of blood plasma proteins (see Blood Plasma). Of the five major types of immunoglobulins, the most abundant are immunoglobulin G, immunoglobulin A, and immunoglobulin M.

Immunoglobulin G (IgG) is in plasma and tissue fluids and is particularly effective against bacteria, viruses, and toxins. It also activates complement.

Immunoglobulin A (IgA) is commonly found in exocrine gland secretions. It is in breast milk, tears, nasal fluid, gastric juice, intestinal juice, bile, and urine.

Immunoglobulin M (IgM) is a type of antibody present in plasma in response to contact with certain antigens in foods or bacteria. The antibodies anti-A and anti-B, described in Blood Donation – Blood Transfusions, are examples of IgM. IgM also activates complement.

Immunoglobulin D (IgD) is found on the surfaces of most B cells, especially those of infants. IgD is important in activating B cells.

Immunoglobulin E (IgE) is found on the surfaces of basophils and mast cells. It is associated with allergic responses.

A newborn does not yet have its own antibodies, but does retain for a while IgG that passed through the placenta from the mother. These maternal antibodies protect the infant against some illnesses to which the mother is immune. As the maternal antibody supply falls, the infant begins to manufacture its own antibodies. The newborn also receives IgA from colostrum, a substance secreted from the mother’s breasts for the first few days after birth, and then from breast milk. Antibodies in colostrum protect against certain digestive and respiratory infections.

Antibody Actions

In general, antibodies react to antigens in three ways. Antibodies directly attack antigens, activate complement, or stimulate localized changes (inflammation) that help prevent the spread of pathogens or cells bearing foreign antigens.

In a direct attack, antibodies combine with antigens, causing them to clump (agglutination) or to form insoluble substances (precipitation). Such actions make it easier for phagocytic cells to recognize and engulf the antigen-bearing agents and eliminate them. In other instances, antibodies cover the toxic portions of antigen molecules and neutralize their effects (neutralization). However, under normal conditions, direct antibody attack is not as important as complement activation in protecting against infection.

When certain IgG or IgM antibodies combine with antigens, they expose reactive sites on antibody molecules. This triggers a series of reactions, leading to activation of the complement proteins, which in turn produce a variety of effects. These include:

  • coating the antigen-antibody complexes (opsonization), making the complexes more susceptible to phagocytosis;
  • attracting macrophages and neutrophils into the region (chemotaxis);
  • rupturing membranes of foreign cells (lysis); agglutination of antigen-bearing cells; and
  • neutralization of viruses by altering their molecular structure, making them harmless.

Other proteins promote inflammation, which helps prevent the spread of infectious agents.

Immune Responses

Activation of B cells or T cells after they first encounter the antigens for which they are specialized to react constitutes a primary immune response. During such a response, plasma cells release antibodies (IgM, followed by IgG) into the lymph. The antibodies are transported to the blood and then throughout the body, where they help destroy antigen bearing agents. Production and release of antibodies continues for several weeks.

Following a primary immune response, some of the B cells produced during proliferation of the clone remain dormant as memory cells. If the same antigen is encountered again, the clones of these memory cells enlarge, and they can respond rapidly by producing IgG to the antigen to which they were previously sensitized. These memory B cells, along with the memory cytotoxic T cells, produce a secondary immune response.

As a result of a primary immune response, detectable concentrations of antibodies usually appear in the blood plasma five to ten days after exposure to antigens. If the same type of antigen is encountered later, a secondary immune response may produce the same antibodies within a day or two. Although newly formed antibodies may persist in the body for only a few months or years, memory cells live much longer.

Naturally acquired active immunity occurs when a person exposed to a pathogen develops a disease. Resistance to that pathogen is the result of a primary immune response.

A vaccine is a preparation that produces artificially acquired active immunity. A vaccine might consist of bacteria or viruses that have been killed or weakened so that they cannot cause a serious infection, or only molecules unique to the pathogens. A vaccine might also be a toxoid, which is a toxin from an infectious organism that has been chemically altered to destroy its dangerous effects. Whatever its composition, a vaccine includes the antigens that stimulate a primary immune response, but does not produce symptoms of disease and the associated infections.

Specific vaccines stimulate active immunity against a variety of diseases, including typhoid fever, cholera, whooping cough, diphtheria, tetanus, polio, chickenpox, measles (rubeola), German measles (rubella), mumps, influenza, hepatitis A, hepatitis B, and bacterial pneumonia. A vaccine has eliminated naturally acquired smallpox from the world.

Autoimmunity

The immune response can turn against the body itself. It may become unable to distinguish a particular self antigen from a nonself antigen, producing  autoantibodies and cytotoxic T cells that attack and damage the body’s tissues and organs. This reaction against self is called autoimmunity.

The specific nature of an autoimmune disorder reflects the cell types that are the target of the immune attack. In type 1 (insulin dependent) diabetes mellitus the target is beta cells in the pancreas. The tissues within the joints are targeted in rheumatoid arthritis. In systemic lupus erythematosus the target is DNA and proteins associated with it in the cell nuclei. About 5% of the population has an autoimmune disorder.

Why might the immune response attack body tissues ?

Perhaps a virus, while replicating inside a human cell, takes proteins from the host cell’s surface and incorporates them onto its own surface. When the immune response “learns” the surface of the virus in order to destroy it, it also learns to attack the human cells that normally bear those particular proteins. Another explanation of autoimmunity is that somehow T cells never learn to distinguish self from nonself. A third possible route of autoimmunity is when a nonself antigen coincidentally resembles a self antigen.

What does low white blood cells mean

Low white blood cells or leucopenia is a range of disorders can cause decreases in white blood cells. Leukopenia is characterized by a reduction in the circulating white blood cell count to < 4000/μL 11. The type of white blood cell decreased is usually the neutrophil. In this case the decrease may be called neutropenia or granulocytopenia. Less commonly, a decrease in lymphocytes (called lymphocytopenia or lymphopenia) may be seen.

Neutropenia

Neutropenia is an abnormally low level of neutrophils. Neutrophils are a common type of white blood cell important to fighting off infections — particularly those caused by bacteria and fungal infections 12. When neutropenia is present, the inflammatory response to such infections is ineffective.

Normal lower limit of the neutrophil count (total white blood cell × % neutrophils and bands) is 1500/μL in whites and is somewhat lower in blacks (about 1200/μL) 12. Neutrophil counts are not as stable as other cell counts and may vary considerably over short periods, depending on many factors such as activity status, anxiety, infections, and drugs. Thus, several measurements may be needed when determining the severity of neutropenia 12.

Severity of neutropenia relates to the relative risk of infection and is classified as follows 12:

  • Mild (1000 to 1500/μL)
  • Moderate (500 to 1000/μL)
  • Severe (<500/μL)

When neutrophil counts fall to <500/μL, endogenous microbial flora (eg, in the mouth or gut) can cause infections. If the count falls to < 200/μL, inflammatory response may be muted and the usual inflammatory findings of leukocytosis or white blood cells in the urine or at the site of infection may not occur. Acute, severe neutropenia, particularly if another factor (eg, cancer) is present, significantly impairs the immune system and can lead to rapidly fatal infections. The integrity of the skin and mucous membranes, the vascular supply to tissue, and the nutritional status of the patient also influence the risk of infections.

The most frequently occurring infections in patients with profound neutropenia are:

  • Cellulitis (infection of subcutaneous connective tissue)
  • Furunculosis (boils on the skin)
  • Pneumonia (lung infection)
  • Septicemia (bacteria in the blood)

Vascular catheters and other puncture sites confer extra risk of skin infections; the most common bacterial causes are coagulase-negative staphylococci and Staphylococcus aureus, but other gram-positive and gram-negative infections also occur. Stomatitis, gingivitis, perirectal inflammation, colitis, sinusitis, paronychia, and otitis media often occur. Patients with prolonged neutropenia after hematopoietic stem cell transplantation or chemotherapy and patients receiving high doses of corticosteroids are predisposed to fungal infections.

Causes of neutropenia

Cancer chemotherapy is probably the most common cause of neutropenia. People with chemotherapy-related neutropenia are prone to infections while they wait for their cell counts to recover.

Neutrophils are manufactured in bone marrow — the spongy tissue inside some of your larger bones. Anything that disrupts neutrophil production can result in neutropenia.

Acute neutropenia (occurring over hours to a few days) can develop as a result of rapid neutrophil use or destruction or due to impaired production.

Chronic neutropenia (lasting months to years) usually arises as a result of reduced production or excessive splenic sequestration.

Neutropenia also may be classified as primary due to an intrinsic defect in marrow myeloid cells or as secondary (due to factors extrinsic to marrow myeloid cells).

Neutropenia can be acquired or intrinsic 13. A decrease in levels of neutrophils on lab tests is due to either decreased production of neutrophils or increased removal from the blood 14.

The following is a list of neutropenia causes due to intrinsic defects in myeloid cells or their precursors:

  • Aplastic anemia
  • Chronic idiopathic neutropenia, including benign neutropenia
  • Cyclic neutropenia
  • Myelodysplasia
  • Neutropenia associated with dysgammaglobulinemia
  • Paroxysmal nocturnal hemoglobinuria
  • Severe congenital neutropenia (Kostmann syndrome)
  • Syndrome-associated neutropenias (eg, cartilage-hair hypoplasia syndrome, dyskeratosis congenita, glycogen storage disease type IB, warts, hypogammaglobulinemia, infections, myelokathexis [WHIM] syndrome, Shwachman-Diamond syndrome)

The following is a list of causes of secondary neutropenias:

  • Alcoholism
  • Autoimmune neutropenia, including chronic secondary neutropenia in AIDS
  • Bone marrow replacement (eg, due to cancer, myelofibrosis, granuloma, or Gaucher cells)
  • Cytotoxic chemotherapy or radiation therapy
  • Drug-induced neutropenia – sulfas or other antibiotics, phenothiazenes, benzodiazepines, antithyroids, anticonvulsants, quinine, quinidine, indomethacin, procainamide, thiazides
  • Folate deficiency, vitamin B12 deficiency, or severe undernutrition
  • Hypersplenism
  • Infection
  • T cell large granular lymphocyte disease

Symptoms and Signs of Neutropenia

Neutropenia is asymptomatic until infection develops. Fever is often the only indication of infection. Typical signs of inflammation (erythema, swelling, pain, infiltrates, leukocytic reaction) may be muted or absent. Focal symptoms (eg, oral ulcers) may develop but are often subtle. Patients with drug-induced neutropenia due to hypersensitivity may have a fever, rash, and lymphadenopathy as a result of the hypersensitivity.

Neutropenia is usually found when your doctor orders tests for a condition you’re already experiencing. It’s rare for neutropenia to be discovered unexpectedly or by chance.

Talk to your doctor about what your test results mean. Neutropenia and results from other tests might indicate the cause of your illness. Or, your doctor may suggest other tests to further check your condition.

Because neutropenia makes you vulnerable to bacterial and fungal infections, your doctor will probably advise certain precautions. These often include wearing a face mask, avoiding anyone with a cold, and washing your hands regularly and thoroughly.

Some patients with chronic benign neutropenia and neutrophil counts < 200/μL do not experience many serious infections. Patients with cyclic neutropenia or severe congenital neutropenia tend to have episodes of oral ulcers, stomatitis, or pharyngitis and lymph node enlargement during severe neutropenia. Pneumonias and septicemia often occur.

Treatment of neutropenia

  • Treatment of associated conditions (eg, infections, stomatitis)
  • Sometimes antibiotic prophylaxis
  • Myeloid growth factors
  • Discontinuation of suspected etiologic agent (eg, drug)
  • Sometimes corticosteroids

Acute neutropenia

Suspected infections are always treated immediately. If fever or hypotension is present, serious infection is assumed, and empiric, high-dose, broad-spectrum antibiotics are given IV. Regimen selection is based on the most likely infecting organisms, the antimicrobial susceptibility of pathogens at that particular institution, and the regimen’s potential toxicity. Because of the risk of creating resistant organisms, vancomycin is used only if gram-positive organisms resistant to other drugs are suspected.

Indwelling vascular catheters can usually remain in place even if bacteremia is suspected or documented, but removal is considered if infections involve S. aureus or Bacillus sp, Corynebacterium sp, or Candida sp or if blood cultures are persistently positive despite appropriate antibiotics. Infections caused by coagulase-negative staphylococci generally resolve with antimicrobial therapy alone. Indwelling Foley catheters can also predispose to infections in these patients, and change or removal of the catheter should be considered for persistent urinary infections.

If cultures are positive, antibiotic therapy is adjusted to the results of sensitivity tests. If a patient defervesces within 72 h, antibiotics are continued for at least 7 days and until the patient has no symptoms or signs of infection. When neutropenia is transient (such as that following myelosuppressive chemotherapy), antibiotic therapy is usually continued until the neutrophil count is >500/μL; however, stopping antimicrobials can be considered in selected patients with persistent neutropenia, especially those in whom symptoms and signs of inflammation have resolved, if cultures remain negative.

Fever that persists > 72 h despite antibiotic therapy suggests a nonbacterial cause, infection with a resistant species, a superinfection with a 2nd bacterial species, inadequate serum or tissue levels of the antibiotics, or localized infection, such as an abscess. Neutropenic patients with persistent fever are reassessed every 2 to 4 days with physical examination, cultures, and chest x-ray. If the patient is well except for the presence of fever, the initial antibiotic regimen can be continued, and drug-induced fever should be considered. If the patient is deteriorating, alteration of the antimicrobial regimen is considered.

Fungal infections are the most likely cause of persistent fevers and deterioration. Antifungal therapy is added empirically if unexplained fever persists after 3 to 4 days of broad-spectrum antibiotic therapy. Selection of the specific antifungal drug (eg, fluconazole, caspofungin, voriconazole, posaconazole) depends on the type of risk (eg, duration and severity of neutropenia, past history of fungal infection, persistent fever despite use of narrower spectrum antifungal drug) and should be guided by an infectious disease specialist. If fever persists after 3 wk of empiric therapy (including 2 wk of antifungal therapy) and the neutropenia has resolved, then stopping all antimicrobials can be considered and the cause of fever reevaluated.

For afebrile patients with neutropenia, antibiotic prophylaxis with fluoroquinolones (levofloxacin, ciprofloxacin) is used in some centers for patients who receive chemotherapy regimens that commonly result in neutrophils ≤ 100/µL for > 7 days.. Prophylaxis is usually started by the treating oncologist. Antibiotics are continued until the neutrophil count increases to > 1500/µL. Also, antifungal therapy can be given for afebrile neutropenic patients at higher risk of fungal infection (eg, after hematopoietic stem cell transplantation, intensive chemotherapy for acute myelogenous leukemia or a myelodysplastic disorder, prior fungal infections). Selection of the specific antifungal drug should be guided by an infectious disease specialist. Antibiotic and antifungal prophylaxis is not routinely recommended for afebrile neutropenic patients without risk factors who are anticipated to remain neutropenic for < 7 days on the basis of their specific chemotherapy regimen.

Myeloid growth factors (ie, granulocyte colony-stimulating factor [G-CSF]) are widely used to increase the neutrophil count and to prevent infections in patients with severe neutropenia (eg, after hematopoietic stem cell transplantation and intensive cancer chemotherapy). They are expensive. However, if the risk of febrile neutropenia is ≥ 30% (as assessed by neutrophil count < 500 μL, presence of infection during a previous cycle of chemotherapy, associated comorbid disease, or age > 75), growth factors are indicated. In general, most clinical benefit occurs when the growth factor is administered beginning about 24 h after completion of chemotherapy. Patients with neutropenia caused by an idiosyncratic drug reaction may also benefit from myeloid growth factors, particularly if a delayed recovery is anticipated. The dose for G-CSF (filgrastim) is 5 to 10 mcg/kg sc once/day, and the dose for pegylated G-CSF (pegfilgrastim) is 6 mg sc once per chemotherapy cycle.

Glucocorticoids, anabolic steroids, and vitamins do not stimulate neutrophil production but can affect distribution and destruction. If acute neutropenia is suspected to be drug- or toxin-induced, all potentially etiologic agents are stopped. If neutropenia develops during treatment with a drug known to induce low counts (eg, chloramphenicol), then switching to an alternative antibiotic may be helpful.

Saline or hydrogen peroxide gargles every few hours, liquid oral rinses (containing viscous lidocaine, diphenhydramine, and liquid antacid), anesthetic lozenges (benzocaine 15 mg q 3 or 4 h), or chlorhexidine mouth rinses (1% solution) bid or tid may relieve the discomfort of stomatitis with oropharyngeal ulcerations. Oral or esophageal candidiasis is treated with nystatin (400,000 to 600,000 units oral rinse qid; swallowed if esophagitis is present), clotrimazole troche (10 mg slowly dissolved in the mouth 5 times a day), or systemic antifungal drugs (eg, fluconazole). A semisolid or liquid diet may be necessary during acute stomatitis or esophagitis, and topical analgesics (eg,viscous lidocaine) may be needed to minimize discomfort.

Chronic neutropenia

Neutrophil production in congenital neutropenia, cyclic neutropenia, and idiopathic neutropenia can be increased with administration of G-CSF 1 to 10 mcg/kg sc once/day. Effectiveness can be maintained with daily or intermittent G-CSF for months or years. Long-term G-CSF has also been used in other patients with chronic neutropenia, including those with myelodysplasia, HIV, and autoimmune disorders. In general, neutrophil counts increase, although clinical benefits are less clear, especially for patients who do not have severe neutropenia. For patients with autoimmune disorders or who have had an organ transplant, cyclosporine can also be beneficial.

In some patients with accelerated neutrophil destruction caused by autoimmune disorders, corticosteroids (generally, prednisone 0.5 to 1.0 mg/kg po once/day) can increase blood neutrophils. This increase often can be maintained with alternate-day G-CSF therapy.

Splenectomy has been used in the past to increase the neutrophil count in some patients with splenomegaly and splenic sequestration of neutrophils (eg, Felty syndrome); however,because growth factors and other newer therapies are often effective, splenectomy should be avoided in most patients. However, splenectomy can be considered for patients with persistent painful splenomegaly or with severe neutropenia (ie, <500/μL) and serious problems with infections in whom other treatments have failed. Patients should be vaccinated against infections caused by Streptococcus pneumoniae, Neisseria meningitidis, and Haemophilus influenzae before splenectomy because splenectomy predisposes patients to infection by encapsulated organisms.

Lymphocytopenia

The normal lymphocyte count in adults is 1000 to 4800/μL; in children < 2 yr, 3000 to 9500/μL. At age 6 yr, the lower limit of normal is 1500/μL.

Lymphocytopenia is a total lymphocyte count of < 1000/μL in adults or < 3000/μL in children < 2 yr 15.

Both B and T cells are present in the peripheral blood; about 75% of the lymphocytes are T cells and 25% B cells. Because lymphocytes account for only 20 to 40% of the total WBC count, lymphocytopenia may go unnoticed when WBC count is checked without a differential.

Almost 65% of blood T cells are CD4+ T-helper cells. Most patients with lymphocytopenia have a reduced absolute number of T cells, particularly in the number of CD4+ T cells. The average number of CD4+ T-helper cells in adult blood is 1100/μL (range, 300 to 1300/μL), and the average number of cells of the other major T-cell subgroup, CD8+ (suppressor) T cells, is 600/μL (range, 100 to 900/μL).

Lymphocytopenia can be 15:

  • Acquired
  • Inherited

Causes of Acquired lymphocytopenia:

  • AIDS
  • Other infectious disorders, including hepatitis, influenza, TB, typhoid fever, and sepsis
  • Dietary deficiency in patients with ethanol abuse, protein-energy undernutrition, or zinc deficiency
  • Protein losing enteropathy
  • Iatrogenic after use of cytotoxic chemotherapy, glucocorticoids, high-dose psoralen and ultraviolet A radiation therapy, lymphocyte antibody therapy, immunosuppressants, radiation therapy, or thoracic duct drainage
  • Systemic disorders with autoimmune features (eg, aplastic anemia, Hodgkin lymphoma, myasthenia gravis, protein-losing enteropathy, RA, chronic kidney disease, sarcoidosis, SLE, thermal injury).

Causes of Inherited lymphocytopenia:

  • Aplasia of lymphopoietic stem cells
  • Ataxia-telangiectasia
  • Cartilage-hair hypoplasia syndrome
  • Idiopathic CD4+ T lymphocytopenia
  • Immunodeficiency with thymoma
  • Severe combined immunodeficiency associated with a defect in the IL-2 receptor gamma-chain, deficiency of adenosine deaminase or purine nucleoside phosphorylase, or an unknown defect
  • Wiskott-Aldrich syndrome

Acquired lymphocytopenia

Acquired lymphocytopenia can occur with a number of other disorders. The most common causes include:

  • Protein-energy undernutrition
  • AIDS and certain other viral infections

Protein-energy undernutrition is the most common cause worldwide.

AIDS is the most common infectious disease causing lymphocytopenia, which arises from destruction of CD4+ T cells infected with HIV. Lymphocytopenia may also reflect impaired lymphocyte production arising from destruction of thymic or lymphoid architecture. In acute viremia due to HIV or other viruses, lymphocytes may undergo accelerated destruction from active infections with the virus, may be trapped in the spleen or lymph nodes, or may migrate to the respiratory tract.

Iatrogenic lymphocytopenia is caused by cytotoxic chemotherapy, radiation therapy, or the administration of antilymphocyte globulin (or other lymphocyte antibodies). Long-term treatment for psoriasis using psoralen and ultraviolet A irradiation may destroy T cells. Glucocorticoids can induce lymphocyte destruction.

Lymphocytopenia may occur with lymphomas, autoimmune diseases such as SLE, rheumatoid arthritis, myasthenia gravis, and protein-losing enteropathy.

Inherited lymphocytopenia

Inherited lymphocytopenia (see Table: Causes of Lymphocytopenia) most commonly occurs in:

  • Severe combined immunodeficiency disorder
  • Wiskott-Aldrich syndrome

It may occur with inherited immunodeficiency disorders and disorders that involve impaired lymphocyte production. Other inherited disorders, such as Wiskott-Aldrich syndrome, adenosine deaminase deficiency, and purine nucleoside phosphorylase deficiency, may involve accelerated T-cell destruction. In many disorders, antibody production is also deficient.

Symptoms and Signs of Lymphocytopenia

Lymphocytopenia per se generally causes no symptoms. However, findings of an associated disorder may include:

  • Absent or diminished tonsils or lymph nodes, indicative of cellular immunodeficiency
  • Skin abnormalities (eg, alopecia, eczema, pyoderma, telangiectasia)
  • Evidence of hematologic disease (eg, pallor, petechiae, jaundice, mouth ulcers)
  • Generalized lymphadenopathy and splenomegaly, which may suggest HIV infection or Hodgkin lymphoma

Lymphocytopenic patients experience recurrent infections or develop infections with unusual organisms. Pneumocystis jirovecii, cytomegalovirus, rubeola, and varicella pneumonias often are fatal. Lymphocytopenia is also a risk factor for the development of cancers and for autoimmune disorders.

Diagnosis of Lymphocytopenia

  • Clinical suspicion (repeated or unusual infections)
  • Complete blood count with differential
  • Measurement of lymphocyte subpopulations and immunoglobulin levels

Lymphocytopenia is suspected in patients with recurrent viral, fungal, or parasitic infections but is usually detected incidentally on a complete blood count. P. jirovecii, cytomegalovirus, rubeola, or varicella pneumonias with lymphocytopenia suggest immunodeficiency. Lymphocyte subpopulations are measured in patients with lymphocytopenia. Measurement of immunoglobulin levels should also be done to evaluate antibody production. Patients with a history of recurrent infections undergo complete laboratory evaluation for immunodeficiency, even if initial screening tests are normal.

Treatment of Lymphocytopenia

  • Treatment of associated infections
  • Treatment of underlying disorder
  • Sometimes IV immune globulin
  • Possibly hematopoietic stem cell transplantation

In acquired lymphocytopenias, lymphocytopenia usually remits with removal of the underlying factor or successful treatment of the underlying disorder. IV immune globulin is indicated if patients have chronic IgG deficiency, lymphocytopenia, and recurrent infections. Hematopoietic stem cell transplantation can be considered for all patients with congenital immunodeficiencies and may be curative.

What does increase white blood cells mean ?

A high white blood cell count is also called leukocytosis. A high white blood cell count is an increase in disease-fighting cells in your blood.

The exact threshold for a high white blood cell count varies from one laboratory to another. The normal range for white blood cell counts changes with age and pregnancy (Table 1).  In general, for nonpregnant adults a count of more than 11,000 per mm3 (11.0 × 109 per L) white blood cells (leukocytes) in a microliter of blood is considered a high white blood cell count 16.

Leukocytosis in the range of approximately 50,000 to 100,000 per mm3 (50.0 to 100.0 × 109 per L) is sometimes referred to as a leukemoid reaction. This level of elevation can occur in some severe infections, such as Clostridium difficile infection, sepsis, organ rejection, or in patients with solid tumors 3. Leukocytosis greater than 100,000 per mm3 is almost always caused by leukemias or myeloproliferative disorders 17.

The most common type of leukocytosis is neutrophilia (an increase in the absolute number of mature neutrophils to greater than 7,000 per mm3 [7.0 × 109 per L]), which can arise from infections, stressful conditions, chronic inflammation, medication use, and other causes (Table 4) 3, 8, 18, 19. Lymphocytosis (when lymphocytes make up more than 40% of the white blood cell count or the absolute count is greater than 4,500 per mm3 [4.5 × 109 per L]) can occur in patients with pertussis, syphilis, viral infections, hypersensitivity reactions, and certain subtypes of leukemia or lymphoma. Lymphocytosis is more likely to be benign in children than in adults 5. Epstein-Barr virus infection, tuberculosis or fungal disease, autoimmune disease, splenectomy, protozoan or rickettsial infections, and malignancy can cause monocytosis (monocytes make up more than 8% of the white blood cell count or the absolute count is greater than 880 per mm3 [0.88 × 109 per L]) 9.

Eosinophilia (eosinophil absolute count greater than 500 per mm3 [0.5 × 109 per L]), although uncommon, may suggest allergic conditions such as asthma, urticaria, atopic dermatitis or eosinophilic esophagitis, drug reactions, dermatologic conditions, malignancies, connective tissue disease, idiopathic hypereosinophilic syndrome, or parasitic infections, including helminths (tissue parasites more than gut-lumen parasites) 20, 21. Isolated basophilia (number of basophils greater than 100 per mm3 [0.1 × 109 per L]) is rare and unlikely to cause leukocytosis in isolation, but it can occur with allergic or inflammatory conditions and chronic myelogenous leukemia 22 (Table 5).

A high white blood cell count is usually found when your doctor orders tests to help diagnose a condition you’re already experiencing. It’s rarely an unexpected finding or simply discovered by chance.

Speak to your doctor about what these results mean. A high white blood cell count, along with results from other tests, might already indicate the cause of your illness. Or your doctor may suggest other tests to further evaluate your condition.

Nonmalignant Causes of of Raised White Blood Cell

A reactive leukocytosis, typically in the range of 11,000 to 30,000 per mm3 (11.0 to 30.0 × 109 per L), can arise from a variety of etiologies. Any source of stress can cause a catecholamine-induced demargination of white blood cells, as well as increased release from the bone marrow storage pool. Examples include surgery, exercise, trauma, burns, and emotional stress.9 One study showed an average increase in white blood cells of 2,770 per mm3 (2.77 × 109 per L) peaking on postoperative day 2 after knee or hip arthroplasty.10 Medications known to increase the white blood cell count include corticosteroids, lithium, colony-stimulating factors, beta agonists, and epinephrine. During the recovery phase after hemorrhage or hemolysis, a rebound leukocytosis can occur.

Leukocytosis is one of the hallmarks of infection. In the acute stage of many bacterial infections, there are primarily mature and immature neutrophils; sometimes, as the infection progresses, there is a shift to lymphocyte predominance. The release of less-mature bands and metamyelocytes into the peripheral circulation results in the so-called “left shift” in the white blood cell differential. Of note, some bacterial infections paradoxically cause neutropenias, such as typhoid fever, rickettsial infections, brucellosis, and dengue 3, 23. Viral infections may cause leukocytosis early in their course, but a sustained leukocytosis is not typical, except for the lymphocytosis in some childhood viral infections.

An elevated white blood cell count is a suggestive, but not definitive, marker of the presence of significant infection. For example, the sensitivity and specificity of an elevated white blood cell count in diagnosing acute appendicitis are 62% and 75%, respectively 24, 25. For diagnosing serious bacterial infections without a source in febrile children, the discriminatory value of leukocytosis is less than that of other biomarkers, such as C-reactive protein or procalcitonin 26. Although a WBC count greater than 12,000 per mm3 (12.0 × 109 per L) is one of the criteria for the systemic inflammatory response syndrome (or sepsis when there is a known infection), leukocytosis alone is a poor predictor of bacteremia and not an indication for obtaining blood cultures 27, 28.

Other acquired causes of leukocytosis include functional asplenia (predominantly lymphocytosis), smoking, and obesity. Patients with a chronic inflammatory condition, such as rheumatoid arthritis, inflammatory bowel disease, or a granulomatous disease, may also exhibit leukocytosis. Genetic causes include hereditary or chronic idiopathic neutrophilia and Down syndrome.

Table 4. Nonmalignant Causes of Neutrophilia

CauseDistinguishing featuresEvaluation
Patient characteristics
Pregnancy, obesity, race, age
Reference appropriate WBC count by age or pregnancy trimester Compare WBC count to recent baseline (if available)
Infection
Fever, system-specific symptoms
Physical examination findings
Obtain system-specific cultures and imaging (e.g., sputum cultures, chest radiography)
Consider empiric antibiotics
Consider use of other biomarkers, such as CRP and procalcitonin
Reactive neutrophilia
Exercise, physical stress (e.g., postsurgical, febrile seizures), emotional stress (e.g., panic attacks), smoking
Confirm with history
Chronic inflammation
Rheumatic disease, inflammatory bowel disease, granulomatous disease, vasculitides, chronic hepatitis
Obtain personal and family medical history
Consider erythrocyte sedimentation rate and CRP levels, specific rheumatology laboratories
Consider subspecialist consultation (e.g., rheumatology, gastroenterology)
Medication induced
Corticosteroids, beta agonists, lithium, epinephrine, colony-stimulating factors
Confirm with history; consider discontinuation of medication, if warranted
Bone marrow stimulation
Hemolytic anemia, immune thrombocytopenia, bone marrow suppression recovery, colony-stimulating factors
Complete blood count differential; compare with baseline values (if available)
Examine peripheral smear
Consider reticulocyte and lactate dehydrogenase levels
Consider flow cytometry, bone marrow examination, hematology/oncology consultation
Splenectomy
History of trauma or sickle cell disease
Confirm with history
Congenital
Hereditary/chronic idiopathic neutrophilia, Down syndrome, leukocyte adhesion deficiency
Obtain family, developmental history
Consider hematology/oncology, genetics, and immunology consultations

Note: After patient characteristics, causes are listed in approximate order of frequency.

CRP = C-reactive protein; WBC = white blood cell.

[Source 29]

Table 5. Selected Conditions Associated with Elevations in Certain White Blood Cell Types

White blood cell lineConditions that typically cause elevations

Basophils

Allergic conditions, leukemias

Eosinophils

Allergic conditions, dermatologic conditions, eosinophilic esophagitis, idiopathic hypereosinophilic syndrome, malignancies, medication reactions, parasitic infections

Lymphocytes

Acute or chronic leukemia, hypersensitivity reaction, infections (viral, pertussis)

Monocytes

Autoimmune disease, infections (Epstein-Barr virus, fungal, protozoan, rickettsial, tuberculosis), splenectomy

Neutrophils

Bone marrow stimulation, chronic inflammation, congenital, infection, medication induced, reactive, splenectomy

[Source 29]

Causes of high white blood cells

A high white blood cell count usually indicates 16:

  • An increased production of white blood cells to fight an infection
  • A reaction to a drug that increases white blood cell production
  • A disease of bone marrow, causing abnormally high production of white blood cells
  • An immune system disorder that increases white blood cell production

Specific causes of a high white blood cell count include 16:

  • Acute lymphocytic leukemia
  • Acute myelogenous leukemia (AML)
  • Allergy, especially severe allergic reactions
  • Chronic lymphocytic leukemia
  • Chronic myelogenous leukemia
  • Drugs, such as corticosteroids and epinephrine
  • Infections, bacterial or viral
  • Myelofibrosis
  • Polycythemia vera
  • Rheumatoid arthritis
  • Smoking
  • Stress, such as severe emotional or physical stress
  • Tuberculosis
  • Whooping cough.

Malignant Causes of Raised White Blood Cells

Leukocytosis may herald a malignant disorder, such as an acute or chronic leukemia or a myeloproliferative disorder, such as polycythemia vera, myelofibrosis, or essential thrombocytosis 30. Many solid tumors may lead to a leukocytosis in the leukemoid range, either through bone marrow involvement or production of granulocyte colony-stimulating or granulocyte-macrophage colony-stimulating factors 31. Chronic leukemias are most commonly diagnosed after incidental findings of leukocytosis on complete blood counts in asymptomatic patients. Patients with features suggestive of hematologic malignancies require prompt referral to a hematologist/oncologist (Table 6) 32.

Table 6. Findings Suggestive of Blood Cancers in the Setting of Leukocytosis

Symptoms

Bruising/bleeding tendency

Fatigue, weakness

Fever > 100.4°F (38°C)

Immunosuppression

Night sweats

Unintentional weight loss

Physical examination findings

Lymphadenopathy

Petechiae

Splenomegaly or hepatomegaly

Laboratory abnormalities

Decreased red blood cell count or hemoglobin/hematocrit levels

Increased or decreased platelet count

Monomorphic lymphocytosis on peripheral smear

Predominantly immature cells on peripheral smear

White blood cell count > 30,000 per mm3 (30.0 × 109 per L), or > 20,000 per mm3 (20.0 × 109 per L) after initial management

[Source 32] References
  1. Maton, D., Hopkins, J., McLaughlin, Ch. W., Johnson, S., Warner, M. Q., LaHart, D., & Wright, J. D., Deep V. Kulkarni (1997). Human Biology and Health. Englewood Cliffs, New Jersey, US: Prentice Hall. ISBN 0-13-981176-1.
  2. LaFleur-Brooks, M. (2008). Exploring Medical Language: A Student-Directed Approach (7th ed.). St. Louis, Missouri, US: Mosby Elsevier. p. 398. ISBN 978-0-323-04950-4.
  3. Cerny J, Rosmarin AG. Why does my patient have leukocytosis? Hematol Oncol Clin North Am. 2012;26(2):303–319, viii.
  4. Hoffman R, Benz EJ Jr, Silberstein LE, Heslop H, Weitz J, Anastasi J. Hematology: Basic Principles and Practice. 6th ed. Philadelphia, Pa.: Elsevier/Saunders; 2013:table 164–20.
  5. Chabot-Richards DS, George TI. Leukocytosis. Int J Lab Hematol. 2014;36(3):279–288.
  6. Lurie S, Rahamim E, Piper I, Golan A, Sadan O. Total and differential leukocyte counts percentiles in normal pregnancy. Eur J Obstet Gynecol Reprod Biol. 2008;136(1):16–19.
  7. Dior UP, Kogan L, Elchalal U, et al. Leukocyte blood count during early puerperium and its relation to puerperal infection. J Matern Fetal Neonatal Med. 2014;27(1):18–23.
  8. Lim EM, Cembrowski G, Cembrowski M, Clarke G. Race-specific WBC and neutrophil count reference intervals. Int J Lab Hematol. 2010;32(6 pt 2):590–597.
  9. Berliner N. Leukocytosis and leukopenia. In: Goldman L, Schafer AI, eds. Goldman’s Cecil Medicine. 24th ed. Philadelphia, Pa.: Elsevier/Saunders; 2012.
  10. White blood cell. Wikipedia. https://en.wikipedia.org/wiki/White_blood_cell
  11. Overview of Leukopenias. Merck Manual. https://www.merckmanuals.com/professional/hematology-and-oncology/leukopenias/overview-of-leukopenias
  12. Neutropenia. Merck Manual. https://www.merckmanuals.com/professional/hematology-and-oncology/leukopenias/neutropenia
  13. Richard A. McPherson; Matthew R. Pincus; Naif Z. Abraham Jr.; et al. (eds.). Henry’s clinical diagnosis and management by laboratory methods (22nd ed.). Philadelphia, PA: Elsevier/Saunders. ISBN 1437709745.
  14. Vinay Kumar; et al. (2010). Robbins and Cotran pathologic basis of disease. (8th ed.). Philadelphia, PA: Saunders/Elsevier. ISBN 1416031219.
  15. Lymphocytopenia. Merck Manual. https://www.merckmanuals.com/professional/hematology-and-oncology/leukopenias/lymphocytopenia
  16. High white blood cell count. Mayo Clinic. http://www.mayoclinic.org/symptoms/high-white-blood-cell-count/basics/definition/sym-20050611
  17. Jain R, Bansal D, Marwaha RK. Hyperleukocytosis: emergency management. Indian J Pediatr. 2013;80(2):144–148.
  18. Iskandar JW, Griffeth B, Sapra M, Singh K, Giugale JM. Panic-attack-induced transient leukocytosis in a healthy male: a case report. Gen Hosp Psychiatry. 2011;33(3):302.e11–302.e12.
  19. Deirmengian GK, Zmistowski B, Jacovides C, O’Neil J, Parvizi J. Leukocytosis is common after total hip and knee arthroplasty. Clin Orthop Relat Res. 2011;469(11):3031–3036.
  20. Ustianowski A, Zumla A. Eosinophilia in the returning traveler. Infect Dis Clin North Am. 2012;26(3):781–789.
  21. Cormier SA, Taranova AG, Bedient C, et al. Pivotal Advance: eosinophil infiltration of solid tumors is an early and persistent inflammatory host response. J Leukoc Biol. 2006;79(6):1131–1139.
  22. Munker R. Leukocytosis, leukopenia, and other reactive changes of myelopoiesis. In: Munker R, Hiller E, Glass J, Paquette R, eds. Modern Hematology: Biology and Clinical Management. 2nd ed. Totowa, N.J.; Humana Press; 2007.
  23. Potts JA, Rothman AL. Clinical and laboratory features that distinguish dengue from other febrile illnesses in endemic populations. Trop Med Int Health. 2008;13(11):1328–1340.
  24. Yu CW, Juan LI, Wu MH, Shen CJ, Wu JY, Lee CC. Systematic review and meta-analysis of the diagnostic accuracy of procalcitonin, C-reactive protein and white blood cell count for suspected acute appendicitis. Br J Surg. 2013;100(3):322–329.
  25. Van den Bruel A, Thompson MJ, Haj-Hassan T, et al. Diagnostic value of laboratory tests in identifying serious infections in febrile children: systematic review. BMJ. 2011;342:d3082.
  26. Yo CH, Hsieh PS, Lee SH, et al. Comparison of the test characteristics of procalcitonin to C-reactive protein and leukocytosis for the detection of serious bacterial infections in children presenting with fever without source: a systematic review and meta-analysis. Ann Emerg Med. 2012;60(5):591–600.
  27. Dellinger RP, Levy MM, Rhodes A, et al.; Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med. 2013;41(2):580–637.
  28. Coburn B, Morris AM, Tomlinson G, Detsky AS. Does this adult patient with suspected bacteremia require blood cultures? [published correction appears in JAMA. 2013;309(4):343]. JAMA. 2012;308(5):502–511.
  29. Evaluation of Patients with Leukocytosis. Am Fam Physician. 2015 Dec 1;92(11):1004-1011. http://www.aafp.org/afp/2015/1201/p1004.html
  30. Davis AS, Viera AJ, Mead MD. Leukemia: an overview for primary care. Am Fam Physician. 2014;89(9):731–738.
  31. Granger JM, Kontoyiannis DP. Etiology and outcome of extreme leukocytosis in 758 nonhematologic cancer patients: a retrospective, single-institution study. Cancer. 2009;115(17):3919–3923.
  32. Racil Z, Buresova L, Brejcha M, et al. Clinical and laboratory features of leukemias at the time of diagnosis: an analysis of 1,004 consecutive patients. Am J Hematol. 2011;86(9):800–803.
read more
12 Body SystemsImmune and Hematology System

What are red blood cells ?

red blood cells

What are red blood cells

Blood transports a variety of materials between interior body cells and those that exchange substances with the external environment. In this way, blood helps maintain stable internal environmental conditions. Blood is composed of formed elements suspended in a fluid extracellular matrix called blood plasma. The “formed elements” include red blood cells, white blood cells, and cell fragments called platelets (Figure 2). Most blood cells form in red marrow within the hollow parts of certain long bones.

Most blood samples are roughly 37% to 49% red blood cells by volume – adult females is 38–46% (average = 42%) and for adult males, it is 40–54% (average = 47). This percentage is called the hematocrit. The white blood cells and platelets account for less than 1% of blood volume. The remaining blood sample, about 55%, is the plasma, a clear, straw-colored liquid. Blood plasma is a complex mixture of water, gases, amino acids, proteins, carbohydrates, lipids, vitamins, hormones, electrolytes, and cellular wastes (see Figure 1).

Blood volume varies with body size, percent adipose tissue, and changes in fluid and electrolyte concentrations. An average-size adult has a blood volume of about 5 liters (5.3 quarts), 4–5 liters in a female and 5–6 liters in a male.

Red blood cell (also called erythrocyte) is biconcave disc without a nucleus. This biconcave shape is an adaptation for transporting the gases oxygen and carbon dioxide. It increases the surface area through which oxygen and carbon dioxide can diffuse into and out of the cell (Figures 4 and 5). The characteristic shape of a red blood cell also places the cell membrane closer to oxygen-carrying hemoglobin (Figure 5) molecules in the cell reducing the distance for diffusion.

Each red blood cell is about one-third hemoglobin by volume. This protein imparts the color of blood. When hemoglobin binds oxygen, the resulting oxyhemoglobin is bright red, and when oxygen is released, the resulting deoxyhemoglobin is darker.

Prolonged oxygen deficiency (hypoxia) causes cyanosis, in which the skin and mucous membranes appear bluish due to an abnormally high blood concentration of deoxyhemoglobin in the superficial blood vessels. Exposure to low temperature may also result in cyanosis by constricting superficial blood vessels. This response to environmental change slows skin blood flow. As a result, more oxygen than usual is removed from the blood flowing through the vessels, increasing the concentration of deoxyhemoglobin.

Note: Blood is a complex mixture of formed elements in a liquid extracellular matrix, called blood plasma. Note that water and proteins account for 99% of the blood plasma.

Figure 1. Blood composition

blood composition

blood compositionblood-composition

Note: Blood consists of a liquid portion called plasma and a solid portion (the formed elements) that includes red blood cells, white blood cells, and platelets. When blood components are separated by centrifugation, the white blood cells and platelets form a thin layer, called the “buffy coat,” between the plasma and the red blood cells, which accounts for about 1% of the total blood volume. Blood cells and platelets can be seen under a light microscope when a blood sample is smeared onto a glass slide.

Red Blood Cell Counts

The number of red blood cells in a microliter (μL or mcL or 1 mm3) of blood is called the red blood cell count (RBCC or RCC). This number varies from time to time even in healthy individuals. However, the typical range for adult males is 4,700,000 to 6,100,000 cells per microliter, and that for adult females is 4,200,000 to 5,400,000 cells per microliter.

The absolute numbers for red blood cell, white blood cell, and platelet counts can vary depending on how they are measured and the instruments used to measure them. For this reason, different sources may present different, but very similar, ranges of normal values.

An increase in the number of circulating red blood cells increases the blood’s oxygen-carrying capacity, much as a decrease in the number of circulating red blood cells decreases the blood’s oxygen-carrying capacity. Changes in this number may affect health. For this reason, red blood cell counts are routinely consulted to help diagnose and evaluate the courses of certain diseases.

Blood Cell Formation

The process of blood cell formation, called hematopoiesis, begins in the yolk sac, which lies outside the human embryo. Later in the fetal development, red blood cells are manufactured (erythropoiesis) in the liver and spleen, and still later they form in bone marrow. After birth, these cells are produced in the red bone marrow.

Bone marrow is a soft, netlike mass of connective tissue within the medullary cavities of long bones, in the irregular spaces of spongy bone, and in the larger central canals of compact bone tissue. It is of two kinds: red and yellow. Red bone marrow functions in the formation of red blood cells (erythrocytes), white blood cells (leukocytes), and blood platelets. The color comes from the oxygen-carrying pigment hemoglobin in the red blood cells.

In an infant, red marrow occupies the cavities of most bones. As a person ages, yellow bone marrow, which stores fat, replaces much of the red marrow. Yellow marrow is not active in blood cell production. In an adult, red marrow is primarily found in the spongy bone of the skull, ribs, breastbone (sternum), collarbones (clavicles), backbones (vertebrae), and hip bones. If the supply of blood cells is deficient, some yellow marrow may become red marrow, which then reverts to yellow marrow when the deficiency is corrected.

Figure 3 illustrates the stages in the formation of red blood cells from hematopoietic stem cells (blood-forming cells), which are also called hemocytoblasts.

Red blood cells have nuclei during their early stages of development but lose their nuclei as the cells mature. Losing the nuclei provides more space for hemoglobin. Because mature red blood cells do not have nuclei, they cannot divide. They use none of the oxygen they carry because they do not have mitochondria. Mature red blood cells produce ATP through glycolysis only.

The average life span of a red blood cell is 120 days. Many of these cells are removed from the circulation each day, and yet the number of cells in the circulating blood remains relatively stable. This observation suggests a homeostatic control of the rate of red blood cell production.

The hormone erythropoietin (EPO) controls the rate of red blood cell formation through negative feedback. The kidneys, and to a lesser extent the liver, release erythropoietin in response to prolonged oxygen deficiency (Figure 6). At high altitudes, for example, where the amount of oxygen in the air is reduced, the blood oxygen level initially decreases. This drop in the blood oxygen level triggers the release of erythropoietin, which travels via the blood to the red bone marrow and stimulates red blood cell production.

After a few days of exposure to high altitudes, many newly formed red blood cells appear in the circulating blood. The increased rate of production continues until the number of erythrocytes in the circulation is sufficient to supply tissues with oxygen. When the availability of oxygen returns to normal, erythropoietin release decreases, and the rate of red blood cell production returns to normal as well. An excessive increase in red blood cells is called polycythemia. This condition increases blood viscosity, slowing blood flow and impairing circulation.

Figure 2. Bone marrow anatomy

bone marrow anatomy

Anatomy of the bone. The bone is made up of compact bone, spongy bone, and bone marrow. Compact bone makes up the outer layer of the bone. Spongy bone is found mostly at the ends of bones and contains red marrow. Bone marrow is found in the center of most bones and has many blood vessels. There are two types of bone marrow: red and yellow. Red marrow contains blood stem cells that can become red blood cells, white blood cells, or platelets. Yellow marrow is made mostly of fat.

Dietary Factors Affecting Red Blood Cell Production

Availability of B-complex vitamins—vitamin B12 and folic acid—significantly influences red blood cell production. Because these vitamins are required for DNA synthesis, they are necessary for the growth and division of cells. Cell division is frequent in blood-forming (hematopoietic) tissue, so this tissue is especially vulnerable to a deficiency of either of these vitamins.

Hemoglobin synthesis and normal red blood cell production also require iron. The small intestine absorbs iron slowly from food. The body reuses much of the iron released by the decomposition of hemoglobin from damaged red blood cells. Nonetheless, insufficient dietary iron can reduce hemoglobin synthesis.

A deficiency of red blood cells or a reduction in the amount of hemoglobin they contain results in a condition called anemia. This reduces the oxygen-carrying capacity of the blood, and the affected person may appear pale and lack energy. A pregnant woman may have a normal number of red blood cells, but she develops a relative anemia because her plasma volume increases due to fluid retention. This shows up as a decreased hematocrit.

In contrast to anemia, the inherited disorder called hemochromatosis results in the absorption of iron in the small intestine at ten times the normal rate. Iron builds up in organs, to toxic levels. Treatment is periodic blood removal, as often as every week.

Figure 3. Blood cell development. A blood stem cell goes through several steps to become a red blood cell, platelet, or white blood cell

blood cell development

Figure 4. Blood cells

blood cells

Note: Blood tissue consists of red blood cells, white blood cells, and platelets suspended in plasma. (a) Idealized representation of a sample of blood. (b) Micrograph of a sample of blood (1,000x).

Figure 5. Red blood cells

red blood cells

What is the function of red blood cells

Red blood cells function is to transports oxygen and carbon dioxide. to body tissues by blood flow via circulatory system. Red blood cells take oxygen from the lungs and release it into the cells or tissues. Lipids and proteins make up the cell membrane of red blood cells. Hemoglobin, an iron containing biomolecule, is the rich component of the cytoplasm of red blood cells mainly responsible for the oxygen binding and red color of the erythrocytes (see Figure 6).

Figure 6. Red blood cell formation

red blood cell formation

Note: Low blood oxygen causes the kidneys and to a lesser degree, the liver to release erythropoietin. Erythropoietin stimulates target cells in the red bone marrow to increase the production of red blood cells, which carry oxygen to tissues.

Destruction of Red Blood Cells

The average life span of red blood cells is about four months (120 days) after which it breaks down. Red blood cells are elastic and flexible, and they readily bend as they pass through small blood vessels. As the cells near the end of their four-month life span, however, they become more fragile. The cells may sustain damage simply passing through capillaries, particularly those in active muscles that must withstand strong forces. Macrophages phagocytize and destroy damaged red blood cells, primarily in the liver and spleen. Macrophages are large, phagocytic, wandering cells. During phagocytosis, the iron from the hemoglobin is retained in the liver and spleen cells and is again used in the formation of red blood cells in the body. About 2-10 million red blood cells are formed and destroyed each second in a normal person.

Hemoglobin molecules liberated from red blood cells break down into their four component polypeptide “globin” chains, each surrounding a heme group. The heme further decomposes into iron and a greenish pigment called biliverdin. The blood may transport the iron, combined with a protein, to the hematopoietic tissue in red bone marrow to be reused in synthesizing new hemoglobin. About 80% of the iron is stored in the liver in the form of an iron-protein complex. Biliverdin eventually is converted to an orange-yellow pigment called bilirubin. Biliverdin and bilirubin are secreted in the bile as bile pigments. Figure 7 summarizes the life cycle of a red blood cell.

In jaundice (yellow discoloration of the skin and the whites of the eyes), accumulation of bilirubin turns the skin and eyes yellowish. Newborns can develop physiologic jaundice a few days after birth. This condition may be the result of immature liver cells that ineffectively secrete bilirubin into the bile. Treatment includes exposure to fluorescent light, which breaks down bilirubin in the tissues, and feedings that promote bowel movements. In hospital nurseries, babies being treated for physiological jaundice lie under “bili lights,” clad only in diapers and protective goggles.

Figure 7. Red blood cell hemoglobin

Hemoglobin

Figure 8. Lifecycle of a red blood cell

lifecycle of red blood cell

What happens when you have low red blood cells (Anemia)

Anemia is a condition in which your blood has a lower than normal number of red blood cells 1.

Anemia also can occur if your red blood cells don’t contain enough hemoglobin. Hemoglobin is an iron-rich protein that gives blood its red color. This protein helps red blood cells carry oxygen from the lungs to the rest of the body.

Anemia has three main causes 1:

  1. Blood loss,
  2. Lack of red blood cell production, or
  3. High rates of red blood cell destruction.

These causes might be the result of diseases, conditions, or other factors.

If you have anemia, your body doesn’t get enough oxygen-rich blood. As a result, you may appear pale, feel tired or weak. You also may have other symptoms, such as shortness of breath, dizziness, or headaches.

Severe or long-lasting anemia can damage your heart, brain, and other organs in your body. Very severe anemia may even cause death 1.

Many types of anemia can be mild, short term, and easily treated. You can even prevent some types with a healthy diet. Other types can be treated with dietary supplements.

However, certain types of anemia can be severe, long lasting, and even life threatening if not diagnosed and treated.

If you have signs or symptoms of anemia, see your doctor to find out whether you have the condition. Treatment will depend on the cause of the anemia and how severe it is.

There are many types of anemia with specific causes and traits.

Some of these include:

  • Aplastic anemia
  • Blood loss anemia
  • Cooley’s anemia
  • Diamond-Blackfan anemia
  • Fanconi anemia
  • Folate- or folic acid-deficiency anemia
  • Hemolytic anemia
  • Iron-deficiency anemia
  • Pernicious anemia
  • Sickle cell anemia
  • Thalassemias; Cooley’s anemia is another name for beta thalassemia major

What Causes Anemia ?

The three main causes of anemia are:

  1. Blood loss.
  2. Lack of red blood cell production.
  3. High rates of red blood cell destruction.

For some people, the condition is caused by more than one of these factors.

Blood Loss

Blood loss is the most common cause of anemia, especially iron-deficiency anemia. Blood loss can be short term or persist over time.

Heavy menstrual periods or bleeding in the digestive or urinary tract can cause blood loss. Surgery, trauma, or cancer also can cause blood loss.

If a lot of blood is lost, the body may lose enough red blood cells to cause anemia.

Lack of Red Blood Cell Production

Both acquired and inherited conditions and factors can prevent your body from making enough red blood cells. “Acquired” means you aren’t born with the condition, but you develop it. “Inherited” means your parents passed the gene for the condition on to you.

Acquired conditions and factors that can lead to anemia include poor diet, abnormal hormone levels, some chronic (ongoing) diseases, and pregnancy.

Aplastic anemia also can prevent your body from making enough red blood cells. This condition can be acquired or inherited.

Diet

A diet that lacks iron, folic acid (folate), or vitamin B12 can prevent your body from making enough red blood cells. Your body also needs small amounts of vitamin C, riboflavin, and copper to make red blood cells.

Conditions that make it hard for your body to absorb nutrients also can prevent your body from making enough red blood cells.

Hormones

Your body needs the hormone erythropoietin to make red blood cells. This hormone stimulates the bone marrow to make these cells. A low level of this hormone can lead to anemia.

Diseases and Disease Treatments

Chronic diseases, like kidney disease and cancer, can make it hard for your body to make enough red blood cells.

Some cancer treatments may damage the bone marrow or damage the red blood cells’ ability to carry oxygen. If the bone marrow is damaged, it can’t make red blood cells fast enough to replace the ones that die or are destroyed.

People who have HIV/AIDS may develop anemia due to infections or medicines used to treat their diseases.

Pregnancy

Anemia can occur during pregnancy due to low levels of iron and folic acid and changes in the blood.

During the first 6 months of pregnancy, the fluid portion of a woman’s blood (the plasma) increases faster than the number of red blood cells. This dilutes the blood and can lead to anemia.

Aplastic Anemia

Some infants are born without the ability to make enough red blood cells. This condition is called aplastic anemia. Infants and children who have aplastic anemia often need blood transfusions to increase the number of red blood cells in their blood.

Acquired conditions or factors, such as certain medicines, toxins, and infectious diseases, also can cause aplastic anemia.

High Rates of Red Blood Cell Destruction

Both acquired and inherited conditions and factors can cause your body to destroy too many red blood cells. One example of an acquired condition is an enlarged or diseased spleen.

The spleen is an organ that removes wornout red blood cells from the body. If the spleen is enlarged or diseased, it may remove more red blood cells than normal, causing anemia.

Examples of inherited conditions that can cause your body to destroy too many red blood cells include sickle cell anemia, thalassemias, and lack of certain enzymes. These conditions create defects in the red blood cells that cause them to die faster than healthy red blood cells.

Hemolytic anemia is another example of a condition in which your body destroys too many red blood cells. Inherited or acquired conditions or factors can cause hemolytic anemia. Examples include immune disorders, infections, certain medicines, or reactions to blood transfusions.

Who Is at Risk for Anemia ?

Anemia is a common condition. It occurs in all age, racial, and ethnic groups. Both men and women can have anemia. However, women of childbearing age are at higher risk for the condition because of blood loss from menstruation.

Anemia can develop during pregnancy due to low levels of iron and folic acid (folate) and changes in the blood. During the first 6 months of pregnancy, the fluid portion of a woman’s blood (the plasma) increases faster than the number of red blood cells. This dilutes the blood and can lead to anemia.

During the first year of life, some babies are at risk for anemia because of iron deficiency. At-risk infants include those who are born too early and infants who are fed breast milk only or formula that isn’t fortified with iron. These infants can develop iron deficiency by 6 months of age.

Infants between 1 and 2 years of age also are at risk for anemia. They may not get enough iron in their diets, especially if they drink a lot of cow’s milk. Cow’s milk is low in the iron needed for growth.

Drinking too much cow’s milk may keep an infant or toddler from eating enough iron-rich foods or absorbing enough iron from foods.

Older adults also are at increased risk for anemia. Researchers continue to study how the condition affects older adults. Many of these people have other medical conditions as well.

Major Risk Factors

Factors that raise your risk for anemia include:

  • A diet that is low in iron, vitamins, or minerals
  • Blood loss from surgery or an injury
  • Long-term or serious illnesses, such as kidney disease, cancer, diabetes, rheumatoid arthritis, HIV/AIDS, inflammatory bowel disease (including Crohn’s disease), liver disease, heart failure, and thyroid disease
  • Long-term infections
  • A family history of inherited anemia, such as sickle cell anemia or thalassemia

What Are the Signs and Symptoms of Anemia ?

The most common symptom of anemia is fatigue (feeling tired or weak). If you have anemia, you may find it hard to find the energy to do normal activities.

Other signs and symptoms of anemia include:

  • Shortness of breath
  • Dizziness
  • Headache
  • Coldness in the hands and feet
  • Pale skin
  • Chest pain

These signs and symptoms can occur because your heart has to work harder to pump oxygen-rich blood through your body.

Mild to moderate anemia may cause very mild symptoms or none at all.

Complications of Anemia

Some people who have anemia may have arrhythmias. Arrhythmias are problems with the rate or rhythm of the heartbeat. Over time, arrhythmias can damage your heart and possibly lead to heart failure.

Anemia also can damage other organs in your body because your blood can’t get enough oxygen to them.

Anemia can weaken people who have cancer or HIV/AIDS. This can make their treatments not work as well.

Anemia also can cause many other health problems. People who have kidney disease and anemia are more likely to have heart problems. With some types of anemia, too little fluid intake or too much loss of fluid in the blood and body can occur. Severe loss of fluid can be life threatening.

How Is Anemia Diagnosed ?

Your doctor will diagnose anemia based on your medical and family histories, a physical exam, and results from tests and procedures.

Because anemia doesn’t always cause symptoms, your doctor may find out you have it while checking for another condition.

Medical and Family Histories

Your doctor may ask whether you have any of the common signs or symptoms of anemia. He or she also may ask whether you’ve had an illness or condition that could cause anemia.

Let your doctor know about any medicines you take, what you typically eat (your diet), and whether you have family members who have anemia or a history of it.

Physical Exam

Your doctor will do a physical exam to find out how severe your anemia is and to check for possible causes. He or she may:

  • Listen to your heart for a rapid or irregular heartbeat
  • Listen to your lungs for rapid or uneven breathing
  • Feel your abdomen to check the size of your liver and spleen

Your doctor also may do a pelvic or rectal exam to check for common sources of blood loss.

Diagnostic Tests and Procedures

You may have various blood tests and other tests or procedures to find out what type of anemia you have and how severe it is.

Complete Blood Count

Often, the first test used to diagnose anemia is a complete blood count (CBC). The CBC measures many parts of your blood.

The test checks your hemoglobin and hematocrit levels. Hemoglobin is the iron-rich protein in red blood cells that carries oxygen to the body. Hematocrit is a measure of how much space red blood cells take up in your blood. A low level of hemoglobin or hematocrit is a sign of anemia.

The normal range of these levels might be lower in certain racial and ethnic populations. Your doctor can explain your test results to you.

The CBC also checks the number of red blood cells, white blood cells, and platelets in your blood. Abnormal results might be a sign of anemia, another blood disorder, an infection, or another condition.

Finally, the CBC looks at mean corpuscular volume (MCV). MCV is a measure of the average size of your red blood cells and a clue as to the cause of your anemia. In iron-deficiency anemia, for example, red blood cells usually are smaller than normal.

Other Tests and Procedures

If the CBC results show that you have anemia, you may need other tests, such as:

  • Hemoglobin electrophoresis. This test looks at the different types of hemoglobin in your blood. The test can help diagnose the type of anemia you have.
  • A reticulocyte count. This test measures the number of young red blood cells in your blood. The test shows whether your bone marrow is making red blood cells at the correct rate.
  • Tests for the level of iron in your blood and body. These tests include serum iron and serum ferritin tests. Transferrin level and total iron-binding capacity tests also measure iron levels.

Because anemia has many causes, you also might be tested for conditions such as kidney failure, lead poisoning (in children), and vitamin deficiencies (lack of vitamins, such as B12 and folic acid).

If your doctor thinks that you have anemia due to internal bleeding, he or she may suggest several tests to look for the source of the bleeding. A test to check the stool for blood might be done in your doctor’s office or at home. Your doctor can give you a kit to help you get a sample at home. He or she will tell you to bring the sample back to the office or send it to a laboratory.

If blood is found in the stool, you may have other tests to find the source of the bleeding. One such test is endoscopy. For this test, a tube with a tiny camera is used to view the lining of the digestive tract.

Your doctor also may want to do bone marrow tests. These tests show whether your bone marrow is healthy and making enough blood cells.

How Is Anemia Treated ?

Treatment for anemia depends on the type, cause, and severity of the condition. Treatments may include dietary changes or supplements, medicines, procedures, or surgery to treat blood loss.

Goals of Treatment

The goal of treatment is to increase the amount of oxygen that your blood can carry. This is done by raising the red blood cell count and/or hemoglobin level. (Hemoglobin is the iron-rich protein in red blood cells that carries oxygen to the body.)

Another goal is to treat the underlying cause of the anemia.

Dietary Changes and Supplements

Low levels of vitamins or iron in the body can cause some types of anemia. These low levels might be the result of a poor diet or certain diseases or conditions.

To raise your vitamin or iron level, your doctor may ask you to change your diet or take vitamin or iron supplements. Common vitamin supplements are vitamin B12 and folic acid (folate). Vitamin C sometimes is given to help the body absorb iron.

Iron

Your body needs iron to make hemoglobin. Your body can more easily absorb iron from meats than from vegetables or other foods. To treat your anemia, your doctor may suggest eating more meat—especially red meat (such as beef or liver), as well as chicken, turkey, pork, fish, and shellfish.

Nonmeat foods that are good sources of iron include:

  • Spinach and other dark green leafy vegetables
  • Tofu
  • Peas; lentils; white, red, and baked beans; soybeans; and chickpeas
  • Dried fruits, such as prunes, raisins, and apricots
  • Prune juice
  • Iron-fortified cereals and breads

You can look at the Nutrition Facts label on packaged foods to find out how much iron the items contain. The amount is given as a percentage of the total amount of iron you need every day.

Iron also is available as a supplement. It’s usually combined with multivitamins and other minerals that help your body absorb iron.

Doctors may recommend iron supplements for premature infants, infants and young children who drink a lot of cow’s milk, and infants who are fed breast milk only or formula that isn’t fortified with iron.

Large amounts of iron can be harmful, so take iron supplements only as your doctor prescribes.

Vitamin B12

Low levels of vitamin B12 can lead to pernicious anemia. This type of anemia often is treated with vitamin B12 supplements.

Good food sources of vitamin B12 include:

  • Breakfast cereals with added vitamin B12
  • Meats such as beef, liver, poultry, and fish
  • Eggs and dairy products (such as milk, yogurt, and cheese)
  • Foods fortified with vitamin B12, such as soy-based beverages and vegetarian burgers

Folic Acid

Folic acid (folate) is a form of vitamin B that’s found in foods. Your body needs folic acid to make and maintain new cells. Folic acid also is very important for pregnant women. It helps them avoid anemia and promotes healthy growth of the fetus.

Good sources of folic acid include:

  • Bread, pasta, and rice with added folic acid
  • Spinach and other dark green leafy vegetables
  • Black-eyed peas and dried beans
  • Beef liver
  • Eggs
  • Bananas, oranges, orange juice, and some other fruits and juices

Vitamin C

Vitamin C helps the body absorb iron. Good sources of vitamin C are vegetables and fruits, especially citrus fruits. Citrus fruits include oranges, grapefruits, tangerines, and similar fruits. Fresh and frozen fruits, vegetables, and juices usually have more vitamin C than canned ones.

If you’re taking medicines, ask your doctor or pharmacist whether you can eat grapefruit or drink grapefruit juice. This fruit can affect the strength of a few medicines and how well they work.

Other fruits rich in vitamin C include kiwi fruit, strawberries, and cantaloupes.

Vegetables rich in vitamin C include broccoli, peppers, Brussels sprouts, tomatoes, cabbage, potatoes, and leafy green vegetables like turnip greens and spinach.

Medicines

Your doctor may prescribe medicines to help your body make more red blood cells or to treat an underlying cause of anemia. Some of these medicines include:

  • Antibiotics to treat infections.
  • Hormones to treat heavy menstrual bleeding in teenaged and adult women.
  • A man-made version of erythropoietin to stimulate your body to make more red blood cells. This hormone has some risks. You and your doctor will decide whether the benefits of this treatment outweigh the risks.
  • Medicines to prevent the body’s immune system from destroying its own red blood cells.
  • Chelation therapy for lead poisoning. Chelation therapy is used mainly in children. This is because children who have iron-deficiency anemia are at increased risk of lead poisoning.

Procedures

If your anemia is severe, your doctor may recommend a medical procedure. Procedures include blood transfusions and blood and marrow stem cell transplants.

Blood Transfusion

A blood transfusion is a safe, common procedure in which blood is given to you through an intravenous (IV) line in one of your blood vessels. Transfusions require careful matching of donated blood with the recipient’s blood.

For more information, see Blood Transfusion topic below.

Blood and Marrow Stem Cell Transplant

A blood and marrow stem cell transplant replaces your faulty stem cells with healthy ones from another person (a donor). Stem cells are made in the bone marrow. They develop into red and white blood cells and platelets.

During the transplant, which is like a blood transfusion, you get donated stem cells through a tube placed in a vein in your chest. Once the stem cells are in your body, they travel to your bone marrow and begin making new blood cells.

Surgery

If you have serious or life-threatening bleeding that’s causing anemia, you may need surgery. For example, you may need surgery to control ongoing bleeding due to a stomach ulcer or colon cancer.

If your body is destroying red blood cells at a high rate, you may need to have your spleen removed. The spleen is an organ that removes wornout red blood cells from the body. An enlarged or diseased spleen may remove more red blood cells than normal, causing anemia.

What Is Iron-Deficiency Anemia ?

Iron-deficiency anemia is a common, easily treated condition that occurs if you don’t have enough iron in your body. Low iron levels usually are due to blood loss, poor diet, or an inability to absorb enough iron from food 2.

Iron-deficiency anemia usually develops over time if your body doesn’t have enough iron to build healthy red blood cells. Without enough iron, your body starts using the iron it has stored. Soon, the stored iron gets used up.

After the stored iron is gone, your body makes fewer red blood cells. The red blood cells it does make have less hemoglobin than normal.

Iron-deficiency anemia can cause fatigue (tiredness), shortness of breath, chest pain, and other symptoms. Severe iron-deficiency anemia can lead to heart problems, infections, problems with growth and development in children, and other complications.

Infants and young children and women are the two groups at highest risk for iron-deficiency anemia.

Doctors usually can successfully treat iron-deficiency anemia. Treatment will depend on the cause and severity of the condition. Treatments may include dietary changes, medicines, and surgery.

Severe iron-deficiency anemia may require treatment in a hospital, blood transfusions, iron injections, or intravenous iron therapy.

What Causes Iron-Deficiency Anemia ?

Not having enough iron in your body causes iron-deficiency anemia. Lack of iron usually is due to blood loss, poor diet, or an inability to absorb enough iron from food.

Blood Loss

When you lose blood, you lose iron. If you don’t have enough iron stored in your body to make up for the lost iron, you’ll develop iron-deficiency anemia.

In women, long or heavy menstrual periods or bleeding fibroids in the uterus may cause low iron levels. Blood loss that occurs during childbirth is another cause of low iron levels in women.

Internal bleeding (bleeding inside the body) also may lead to iron-deficiency anemia. This type of blood loss isn’t always obvious, and it may occur slowly. Some causes of internal bleeding are:

  • A bleeding ulcer, colon polyp, or colon cancer
  • Regular use of aspirin or other pain medicines, such as nonsteroidal anti-inflammatory drugs (for example, ibuprofen and naproxen)
  • Urinary tract bleeding

Blood loss from severe injuries, surgery, or frequent blood drawings also can cause iron-deficiency anemia.

Poor Diet

The best sources of iron are meat, poultry, fish, and iron-fortified foods (foods that have iron added). If you don’t eat these foods regularly, or if you don’t take an iron supplement, you’re more likely to develop iron-deficiency anemia.

Vegetarian diets can provide enough iron if you eat the right foods. For example, good nonmeat sources of iron include iron-fortified breads and cereals, beans, tofu, dried fruits, and spinach and other dark green leafy vegetables.

During some stages of life, such as pregnancy and childhood, it may be hard to get enough iron in your diet. This is because your need for iron increases during these times of growth and development.

Inability To Absorb Enough Iron

Even if you have enough iron in your diet, your body may not be able to absorb it. This can happen if you have intestinal surgery (such as gastric bypass) or a disease of the intestine (such as Crohn’s disease or celiac disease).

Prescription medicines that reduce acid in the stomach also can interfere with iron absorption.

Who Is at Risk for Iron-Deficiency Anemia ?

  • Infants and Young Children

Infants and young children need a lot of iron to grow and develop. The iron that full-term infants have stored in their bodies is used up in the first 4 to 6 months of life.

Premature and low-birth-weight babies (weighing less than 5.5 pounds) are at even greater risk for iron-deficiency anemia. These babies don’t have as much iron stored in their bodies as larger, full-term infants.

Iron-fortified baby food or iron supplements, when used properly, can help prevent iron-deficiency anemia in infants and young children. Talk with your child’s doctor about your child’s diet.

Young children who drink a lot of cow’s milk may be at risk for iron-deficiency anemia. Milk is low in iron, and too much milk may take the place of iron-rich foods in the diet. Too much milk also may prevent children’s bodies from absorbing iron from other foods.

Children who have lead in their blood also may be at risk for iron-deficiency anemia. Lead can interfere with the body’s ability to make hemoglobin. Lead may get into the body from breathing in lead dust, eating lead in paint or soil, or drinking water that contains lead.

  • Teens

Teens are at risk for iron-deficiency anemia if they’re underweight or have chronic (ongoing) illnesses. Teenage girls who have heavy periods also are at increased risk for the condition.

  • Women

Women of childbearing age are at higher risk for iron-deficiency anemia because of blood loss during their monthly periods. About 1 in 5 women of childbearing age has iron-deficiency anemia.

Pregnant women also are at higher risk for the condition because they need twice as much iron as usual. The extra iron is needed for increased blood volume and for the fetus’ growth.

About half of all pregnant women develop iron-deficiency anemia. The condition can increase a pregnant woman’s risk for a premature or low-birth-weight baby.

  • Adults Who Have Internal Bleeding

Adults who have internal bleeding, such as intestinal bleeding, can develop iron-deficiency anemia due to blood loss. Certain conditions, such as colon cancer and bleeding ulcers, can cause blood loss. Some medicines, such as aspirin, also can cause internal bleeding.

  • Other At-Risk Groups

People who get kidney dialysis treatment may develop iron-deficiency anemia. This is because blood is lost during dialysis. Also, the kidneys are no longer able to make enough of a hormone that the body needs to produce red blood cells.

People who have gastric bypass surgery also may develop iron-deficiency anemia. This type of surgery can prevent the body from absorbing enough iron.

Certain eating patterns or habits may put you at higher risk for iron-deficiency anemia. This can happen if you:

  • Follow a diet that excludes meat and fish, which are the best sources of iron. However, vegetarian diets can provide enough iron if you eat the right foods. For example, good nonmeat sources of iron include iron-fortified breads and cereals, beans, tofu, dried fruits, and spinach and other dark green leafy vegetables.
  • Eat poorly because of money, social, health, or other problems.
  • Follow a very low-fat diet over a long time. Some higher fat foods, like meat, are the best sources of iron.
  • Follow a high-fiber diet. Large amounts of fiber can slow the absorption of iron.

What Are the Signs and Symptoms of Iron-Deficiency Anemia ?

The signs and symptoms of iron-deficiency anemia depend on its severity. Mild to moderate iron-deficiency anemia may have no signs or symptoms.

When signs and symptoms do occur, they can range from mild to severe. Many of the signs and symptoms of iron-deficiency anemia apply to all types of anemia.

  • Signs and Symptoms of Anemia

The most common symptom of all types of anemia is fatigue (tiredness). Fatigue occurs because your body doesn’t have enough red blood cells to carry oxygen to its many parts.

Also, the red blood cells your body makes have less hemoglobin than normal. Hemoglobin is an iron-rich protein in red blood cells. It helps red blood cells carry oxygen from the lungs to the rest of the body.

Anemia also can cause shortness of breath, dizziness, headache, coldness in your hands and feet, pale skin, chest pain, weakness, and fatigue (tiredness).

If you don’t have enough hemoglobin-carrying red blood cells, your heart has to work harder to move oxygen-rich blood through your body. This can lead to irregular heartbeats called arrhythmias, a heart murmur, an enlarged heart, or even heart failure.

In infants and young children, signs of anemia include poor appetite, slowed growth and development, and behavioral problems.

  • Signs and Symptoms of Iron Deficiency

Signs and symptoms of iron deficiency may include brittle nails, swelling or soreness of the tongue, cracks in the sides of the mouth, an enlarged spleen, and frequent infections.

People who have iron-deficiency anemia may have an unusual craving for nonfood items, such as ice, dirt, paint, or starch. This craving is called pica.

Some people who have iron-deficiency anemia develop restless legs syndrome. Restless legs syndrome is a disorder that causes a strong urge to move the legs. This urge to move often occurs with strange and unpleasant feelings in the legs. People who have restless legs syndrome often have a hard time sleeping.

Iron-deficiency anemia can put children at greater risk for lead poisoning and infections.

Some signs and symptoms of iron-deficiency anemia are related to the condition’s causes. For example, a sign of intestinal bleeding is bright red blood in the stools or black, tarry-looking stools.

Very heavy menstrual bleeding, long periods, or other vaginal bleeding may suggest that a woman is at risk for iron-deficiency anemia.

How Is Iron-Deficiency Anemia Diagnosed ?

Your doctor will diagnose iron-deficiency anemia based on your medical history, a physical exam, and the results from tests and procedures.

Once your doctor knows the cause and severity of the condition, he or she can create a treatment plan for you.

Mild to moderate iron-deficiency anemia may have no signs or symptoms. Thus, you may not know you have it unless your doctor discovers it from a screening test or while checking for other problems.

  • Specialists Involved

Primary care doctors often diagnose and treat iron-deficiency anemia. These doctors include pediatricians, family doctors, gynecologists/obstetricians, and internal medicine specialists.

A hematologist (a blood disease specialist), a gastroenterologist (a digestive system specialist), and other specialists also may help treat iron-deficiency anemia.

  • Medical History

Your doctor will ask about your signs and symptoms and any past problems you’ve had with anemia or low iron. He or she also may ask about your diet and whether you’re taking any medicines.

If you’re a woman, your doctor may ask whether you might be pregnant.

Physical Exam

Your doctor will do a physical exam to look for signs of iron-deficiency anemia. He or she may:

  • Look at your skin, gums, and nail beds to see whether they’re pale
  • Listen to your heart for rapid or irregular heartbeats
  • Listen to your lungs for rapid or uneven breathing
  • Feel your abdomen to check the size of your liver and spleen
  • Do a pelvic and rectal exam to check for internal bleeding

Diagnostic Tests and Procedures

Many tests and procedures are used to diagnose iron-deficiency anemia. They can help confirm a diagnosis, look for a cause, and find out how severe the condition is.

Complete Blood Count

Often, the first test used to diagnose anemia is a complete blood count (CBC). The CBC measures many parts of your blood.

This test checks your hemoglobin and hematocrit levels. Hemoglobin is an iron-rich protein in red blood cells that carries oxygen to the body. Hematocrit is a measure of how much space red blood cells take up in your blood. A low level of hemoglobin or hematocrit is a sign of anemia.

The normal range of these levels varies in certain racial and ethnic populations. Your doctor can explain your test results to you.

The CBC also checks the number of red blood cells, white blood cells, and platelets in your blood. Abnormal results may be a sign of infection, a blood disorder, or another condition.

Finally, the CBC looks at mean corpuscular volume (MCV). MCV is a measure of the average size of your red blood cells. The results may be a clue as to the cause of your anemia. In iron-deficiency anemia, for example, red blood cells usually are smaller than normal.

Other Blood Tests

If the CBC results confirm you have anemia, you may need other blood tests to find out what’s causing the condition, how severe it is, and the best way to treat it.

Reticulocyte count. This test measures the number of reticulocytes in your blood. Reticulocytes are young, immature red blood cells. Over time, reticulocytes become mature red blood cells that carry oxygen throughout your body.

A reticulocyte count shows whether your bone marrow is making red blood cells at the correct rate.

Peripheral smear. For this test, a sample of your blood is examined under a microscope. If you have iron-deficiency anemia, your red blood cells will look smaller and paler than normal.

Tests to measure iron levels. These tests can show how much iron has been used from your body’s stored iron. Tests to measure iron levels include:

  • Serum iron. This test measures the amount of iron in your blood. The level of iron in your blood may be normal even if the total amount of iron in your body is low. For this reason, other iron tests also are done.
  • Serum ferritin. Ferritin is a protein that helps store iron in your body. A measure of this protein helps your doctor find out how much of your body’s stored iron has been used.
  • Transferrin level, or total iron-binding capacity. Transferrin is a protein that carries iron in your blood. Total iron-binding capacity measures how much of the transferrin in your blood isn’t carrying iron. If you have iron-deficiency anemia, you’ll have a high level of transferrin that has no iron.

Other tests. Your doctor also may recommend tests to check your hormone levels, especially your thyroid hormone. You also may have a blood test for a chemical called erythrocyte protoporphyrin. This chemical is a building block for hemoglobin.

Children also may be tested for the level of lead in their blood. Lead can make it hard for the body to produce hemoglobin.

Tests and Procedures for Gastrointestinal Blood Loss

To check whether internal bleeding is causing your iron-deficiency anemia, your doctor may suggest a fecal occult blood test. This test looks for blood in the stools and can detect bleeding in the intestines.

If the test finds blood, you may have other tests and procedures to find the exact spot of the bleeding. These tests and procedures may look for bleeding in the stomach, upper intestines, colon, or pelvic organs.

How Is Iron-Deficiency Anemia Treated ?

Treatment for iron-deficiency anemia will depend on its cause and severity. Treatments may include dietary changes and supplements, medicines, and surgery.

Severe iron-deficiency anemia may require a blood transfusion, iron injections, or intravenous (IV) iron therapy. Treatment may need to be done in a hospital.

The goals of treating iron-deficiency anemia are to treat its underlying cause and restore normal levels of red blood cells, hemoglobin, and iron.

Dietary Changes and Supplements

  • Iron

You may need iron supplements to build up your iron levels as quickly as possible. Iron supplements can correct low iron levels within months. Supplements come in pill form or in drops for children.

Large amounts of iron can be harmful, so take iron supplements only as your doctor prescribes. Keep iron supplements out of reach from children. This will prevent them from taking an overdose of iron.

Iron supplements can cause side effects, such as dark stools, stomach irritation, and heartburn. Iron also can cause constipation, so your doctor may suggest that you use a stool softener.

Your doctor may advise you to eat more foods that are rich in iron. The best source of iron is red meat, especially beef and liver. Chicken, turkey, pork, fish, and shellfish also are good sources of iron.

The body tends to absorb iron from meat better than iron from nonmeat foods. However, some nonmeat foods also can help you raise your iron levels. Examples of nonmeat foods that are good sources of iron include:

  • Iron-fortified breads and cereals
  • Peas; lentils; white, red, and baked beans; soybeans; and chickpeas
  • Tofu
  • Dried fruits, such as prunes, raisins, and apricots
  • Spinach and other dark green leafy vegetables
  • Prune juice

The Nutrition Facts labels on packaged foods will show how much iron the items contain. The amount is given as a percentage of the total amount of iron you need every day.

  • Vitamin C

Vitamin C helps the body absorb iron. Good sources of vitamin C are vegetables and fruits, especially citrus fruits. Citrus fruits include oranges, grapefruits, tangerines, and similar fruits. Fresh and frozen fruits, vegetables, and juices usually have more vitamin C than canned ones.

If you’re taking medicines, ask your doctor or pharmacist whether you can eat grapefruit or drink grapefruit juice. Grapefruit can affect the strength of a few medicines and how well they work.

Other fruits rich in vitamin C include kiwi fruit, strawberries, and cantaloupes.

Vegetables rich in vitamin C include broccoli, peppers, Brussels sprouts, tomatoes, cabbage, potatoes, and leafy green vegetables like turnip greens and spinach.
Treatment To Stop Bleeding

If blood loss is causing iron-deficiency anemia, treatment will depend on the cause of the bleeding. For example, if you have a bleeding ulcer, your doctor may prescribe antibiotics and other medicines to treat the ulcer.

If a polyp or cancerous tumor in your intestine is causing bleeding, you may need surgery to remove the growth.

If you have heavy menstrual flow, your doctor may prescribe birth control pills to help reduce your monthly blood flow. In some cases, surgery may be advised.

Treatments for Severe Iron-Deficiency Anemia

  • Blood Transfusion

If your iron-deficiency anemia is severe, you may get a transfusion of red blood cells. A blood transfusion is a safe, common procedure in which blood is given to you through an IV line in one of your blood vessels. A transfusion requires careful matching of donated blood with the recipient’s blood.

A transfusion of red blood cells will treat your anemia right away. The red blood cells also give a source of iron that your body can reuse. However, a blood transfusion is only a short-term treatment. Your doctor will need to find and treat the cause of your anemia.

Blood transfusions are usually reserved for people whose anemia puts them at a higher risk for heart problems or other severe health issues.

  • Iron Therapy

If you have severe anemia, your doctor may recommend iron therapy. For this treatment, iron is injected into a muscle or an IV line in one of your blood vessels.

IV iron therapy presents some safety concerns. It must be done in a hospital or clinic by experienced staff. Iron therapy usually is given to people who need iron long-term but can’t take iron supplements by mouth. This therapy also is given to people who need immediate treatment for iron-deficiency anemia.

What Is Pernicious Anemia ?

Pernicious anemia is a condition in which the body can’t make enough healthy red blood cells because it doesn’t have enough vitamin B12 3. Without enough vitamin B12, your red blood cells don’t divide normally and are too large. They may have trouble getting out of the bone marrow—a sponge-like tissue inside the bones where blood cells are made.

Pernicious anemia is one of two major types of “macrocystic” or “megaloblastic” (large red blood cell) anemia. These terms refer to anemia in which the red blood cells are larger than normal. The other major type of macrocystic anemia is caused by folic acid deficiency.

Rarely, children are born with an inherited disorder that prevents their bodies from making intrinsic factor. This disorder is called congenital pernicious anemia.

Vitamin B12 deficiency also is called cobalamin deficiency and combined systems disease.

The term “pernicious” means “deadly.” The condition is called pernicious anemia because it often was fatal in the past, before vitamin B12 treatments were available. Now, pernicious anemia usually is easy to treat with vitamin B12 pills or shots.

Vitamin B12 is a nutrient found in some foods. The body needs this nutrient to make healthy red blood cells and to keep its nervous system working properly.

People who have pernicious anemia can’t absorb enough vitamin B12 from food. This is because they lack intrinsic factor, a protein made in the stomach 3. A lack of this protein leads to vitamin B12 deficiency.

Other conditions and factors also can cause vitamin B12 deficiency. Examples include infections, surgery, medicines, and diet. Technically, the term “pernicious anemia” refers to vitamin B12 deficiency due to a lack of intrinsic factor 3. Often though, vitamin B12 deficiency due to other causes also is called pernicious anemia.

Without enough red blood cells to carry oxygen to your body, you may feel tired and weak. Severe or long-lasting pernicious anemia can damage the heart, brain, and other organs in the body 3.

Pernicious anemia also can cause other problems, such as nerve damage, neurological problems (such as memory loss), and digestive tract problems. People who have pernicious anemia also may be at higher risk for weakened bone strength and stomach cancer 3.

With ongoing care and proper treatment, most people who have pernicious anemia can recover, feel well, and live normal lives.

Without treatment, pernicious anemia can lead to serious problems with the heart, nerves, and other parts of the body. Some of these problems may be permanent.

What Causes Pernicious Anemia ?

Pernicious anemia is caused by a lack of intrinsic factor or other causes, such as infections, surgery, medicines, or diet.

Lack of Intrinsic Factor

Intrinsic factor is a protein made in the stomach. It helps your body absorb vitamin B12. In some people, an autoimmune response causes a lack of intrinsic factor.

An autoimmune response occurs if the body’s immune system makes antibodies (proteins) that mistakenly attack and damage the body’s tissues or cells.

In pernicious anemia, the body makes antibodies that attack and destroy the parietal cells. These cells line the stomach and make intrinsic factor. Why this autoimmune response occurs isn’t known.

As a result of this attack, the stomach stops making intrinsic factor. Without intrinsic factor, your body can’t move vitamin B12 through the small intestine, where it’s absorbed. This leads to vitamin B12 deficiency.

A lack of intrinsic factor also can occur if you’ve had part or all of your stomach surgically removed. This type of surgery reduces the number of parietal cells available to make intrinsic factor.

Rarely, children are born with an inherited disorder that prevents their bodies from making intrinsic factor. This disorder is called congenital pernicious anemia.

Other Causes

Pernicious anemia also has other causes, besides a lack of intrinsic factor. Malabsorption in the small intestine and a diet lacking vitamin B12 both can lead to pernicious anemia.

Malabsorption in the Small Intestine

Sometimes pernicious anemia occurs because the body’s small intestine can’t properly absorb vitamin B12. This may be the result of:

  • Too many of the wrong kind of bacteria in the small intestine. This is a common cause of pernicious anemia in older adults. The bacteria use up the available vitamin B12 before the small intestine can absorb it.
  • Diseases that interfere with vitamin B12 absorption. One example is celiac disease. This is a genetic disorder in which your body can’t tolerate a protein called gluten. Another example is Crohn’s disease, an inflammatory bowel disease. HIV also may interfere with vitamin B12 absorption.
  • Certain medicines that alter bacterial growth or prevent the small intestine from properly absorbing vitamin B12. Examples include antibiotics and certain diabetes and seizure medicines.
  • Surgical removal of part or all of the small intestine.
  • A tapeworm infection. The tapeworm feeds off of the vitamin B12. Eating undercooked, infected fish may cause this type of infection.
Diet Lacking Vitamin B12

Some people get pernicious anemia because they don’t have enough vitamin B12 in their diets. This cause of pernicious anemia is less common than other causes.

Good food sources of vitamin B12 include:

  • Breakfast cereals with added vitamin B12
  • Meats such as beef, liver, poultry, and fish
  • Eggs and dairy products (such as milk, yogurt, and cheese)
  • Foods fortified with vitamin B12, such as soy-based beverages and vegetarian burgers

Strict vegetarians who don’t eat any animal or dairy products and don’t take a vitamin B12 supplement are at risk for pernicious anemia.

Breastfed infants of strict vegetarian mothers also are at risk for pernicious anemia. These infants can develop anemia within months of being born. This is because they haven’t had enough time to store vitamin B12 in their bodies. Doctors treat these infants with vitamin B12 supplements.

Other groups, such as the elderly and people who suffer from alcoholism, also may be at risk for pernicious anemia. These people may not get the proper nutrients in their diets.

Who Is at Risk for Pernicious Anemia ?

Pernicious anemia is more common in people of Northern European and African descent than in other ethnic groups.

Older people also are at higher risk for the condition. This is mainly due to a lack of stomach acid and intrinsic factor, which prevents the small intestine from absorbing vitamin B12. As people grow older, they tend to make less stomach acid.

Pernicious anemia also can occur in younger people and other populations. You’re at higher risk for pernicious anemia if you:

  • Have a family history of the condition.
  • Have had part or all of your stomach surgically removed. The stomach makes intrinsic factor. This protein helps your body absorb vitamin B12.
  • Have an autoimmune disorder that involves the endocrine glands, such as Addison’s disease, type 1 diabetes, Graves’ disease, or vitiligo. Research suggests a link may exist between these autoimmune disorders and pernicious anemia that’s caused by an autoimmune response.
  • Have had part or all of your small intestine surgically removed. The small intestine is where vitamin B12 is absorbed.
  • Have certain intestinal diseases or other disorders that may prevent your body from properly absorbing vitamin B12. Examples include Crohn’s disease, intestinal infections, and HIV.
  • Take medicines that prevent your body from properly absorbing vitamin B12. Examples of such medicines include antibiotics and certain seizure medicines.
  • Are a strict vegetarian who doesn’t eat any animal or dairy products and doesn’t take a vitamin B12 supplement, or if you eat poorly overall.

What Are the Signs and Symptoms of Pernicious Anemia ?

A lack of vitamin B12 (vitamin B12 deficiency) causes the signs and symptoms of pernicious anemia. Without enough vitamin B12, your body can’t make enough healthy red blood cells, which causes anemia.

Some of the signs and symptoms of pernicious anemia apply to all types of anemia. Other signs and symptoms are specific to a lack of vitamin B12.

  • Signs and Symptoms of Anemia

The most common symptom of all types of anemia is fatigue (tiredness). Fatigue occurs because your body doesn’t have enough red blood cells to carry oxygen to its various parts.

A low red blood cell count also can cause shortness of breath, dizziness, headache, coldness in your hands and feet, pale or yellowish skin, and chest pain.

A lack of red blood cells also means that your heart has to work harder to move oxygen-rich blood through your body. This can lead to irregular heartbeats called arrhythmias, heart murmur, an enlarged heart, or even heart failure.

  • Signs and Symptoms of Vitamin B12 Deficiency

Vitamin B12 deficiency may lead to nerve damage. This can cause tingling and numbness in your hands and feet, muscle weakness, and loss of reflexes. You also may feel unsteady, lose your balance, and have trouble walking. Vitamin B12 deficiency can cause weakened bones and may lead to hip fractures.

Severe vitamin B12 deficiency can cause neurological problems, such as confusion, dementia, depression, and memory loss.

Other symptoms of vitamin B12 deficiency involve the digestive tract. These symptoms include nausea (feeling sick to your stomach) and vomiting, heartburn, abdominal bloating and gas, constipation or diarrhea, loss of appetite, and weight loss. An enlarged liver is another symptom.

A smooth, thick, red tongue also is a sign of vitamin B12 deficiency and pernicious anemia.

Infants who have vitamin B12 deficiency may have poor reflexes or unusual movements, such as face tremors. They may have trouble feeding due to tongue and throat problems. They also may be irritable. If vitamin B12 deficiency isn’t treated, these infants may have permanent growth problems.

How Is Pernicious Anemia Diagnosed ?

Your doctor will diagnose pernicious anemia based on your medical and family histories, a physical exam, and test results.

Your doctor will want to find out whether the condition is due to a lack of intrinsic factor or another cause. He or she also will want to find out the severity of the condition, so it can be properly treated.

Specialists Involved

Primary care doctors—such as family doctors, internists, and pediatricians (doctors who treat children)—often diagnose and treat pernicious anemia. Other kinds of doctors also may be involved, including:

  • A neurologist (nervous system specialist)
  • A cardiologist (heart specialist)
  • A hematologist (blood disease specialist)
  • A gastroenterologist (digestive tract specialist)

Medical and Family Histories

Your doctor may ask about your signs and symptoms. He or she also may ask:

  • Whether you’ve had any stomach or intestinal surgeries
  • Whether you have any digestive disorders, such as celiac disease or Crohn’s disease
  • About your diet and any medicines you take
  • Whether you have a family history of anemia or pernicious anemia
  • Whether you have a family history of autoimmune disorders (such as Addison’s disease, type 1 diabetes, Graves’ disease, or vitiligo). Research suggests a link may exist between these autoimmune disorders and pernicious anemia that’s caused by an autoimmune response.

Physical Exam

During the physical exam, your doctor may check for pale or yellowish skin and an enlarged liver. He or she may listen to your heart for rapid or irregular heartbeats or a heart murmur.

Your doctor also may check for signs of nerve damage. He or she may want to see how well your muscles, eyes, senses, and reflexes work. Your doctor may ask questions or do tests to check your mental status, coordination, and ability to walk.

Diagnostic Tests and Procedures

Blood tests and procedures can help diagnose pernicious anemia and find out what’s causing it.

  • Complete Blood Count

Often, the first test used to diagnose many types of anemia is a complete blood count (CBC). This test measures many parts of your blood. For this test, a small amount of blood is drawn from a vein (usually in your arm) using a needle.

A CBC checks your hemoglobin and hematocrit levels. Hemoglobin is an iron-rich protein that helps red blood cells carry oxygen from the lungs to the rest of the body. Hematocrit is a measure of how much space red blood cells take up in your blood. A low level of hemoglobin or hematocrit is a sign of anemia.

The normal range of these levels may be lower in certain racial and ethnic populations. Your doctor can explain your test results to you.

The CBC also checks the number of red blood cells, white blood cells, and platelets in your blood. Abnormal results may be a sign of anemia, another blood disorder, an infection, or another condition.

Finally, the CBC looks at mean corpuscular volume (MCV). MCV is a measure of the average size of your red blood cells. MCV can be a clue as to what’s causing your anemia. In pernicious anemia, the red blood cells tend to be larger than normal.

  • Other Blood Tests

If the CBC results confirm that you have anemia, you may need other blood tests to find out what type of anemia you have.

A reticulocyte count measures the number of young red blood cells in your blood. The test shows whether your bone marrow is making red blood cells at the correct rate. People who have pernicious anemia have low reticulocyte counts.

Serum folate, iron, and iron-binding capacity tests also can help show whether you have pernicious anemia or another type of anemia.

Another common test, called the Combined Binding Luminescence Test, sometimes gives false results. Scientists are working to develop a more reliable test.

Your doctor may recommend other blood tests to check:

  • Your vitamin B12 level. A low level of vitamin B12 in the blood indicates pernicious anemia. However, a falsely normal or high value of vitamin B12 in the blood may occur if antibodies interfere with the test.
  • Your homocysteine and methylmalonic acid levels. High levels of these substances in your body are a sign of pernicious anemia.
  • For intrinsic factor antibodies and parietal cell antibodies. These antibodies also are a sign of pernicious anemia.

Bone Marrow Tests

Bone marrow tests can show whether your bone marrow is healthy and making enough red blood cells. The two bone marrow tests are aspiration and biopsy.

For aspiration, your doctor removes a small amount of fluid bone marrow through a needle. For a biopsy, your doctor removes a small amount of bone marrow tissue through a larger needle. The samples are then examined under a microscope.

In pernicious anemia, the bone marrow cells that turn into blood cells are larger than normal.

How Is Pernicious Anemia Treated ?

Doctors treat pernicious anemia by replacing the missing vitamin B12 in the body. People who have pernicious anemia may need lifelong treatment.

The goals of treating pernicious anemia include:

  • Preventing or treating the anemia and its signs and symptoms
  • Preventing or managing complications, such as heart and nerve damage
  • Treating the cause of the pernicious anemia (if a cause can be found)

Specific Types of Treatment

Pernicious anemia usually is easy to treat with vitamin B12 shots or pills.

If you have severe pernicious anemia, your doctor may recommend shots first. Shots usually are given in a muscle every day or every week until the level of vitamin B12 in your blood increases. After your vitamin B12 blood level returns to normal, you may get a shot only once a month.

For less severe pernicious anemia, your doctor may recommend large doses of vitamin B12 pills. A vitamin B12 nose gel and spray also are available. These products may be useful for people who have trouble swallowing pills, such as older people who have had strokes.

Your signs and symptoms may begin to improve within a few days after you start treatment. Your doctor may advise you to limit your physical activity until your condition improves.

If your pernicious anemia is caused by something other than a lack of intrinsic factor, you may get treatment for the cause (if a cause can be found). For example, your doctor may prescribe medicines to treat a condition that prevents your body from absorbing vitamin B12.

If medicines are the cause of your pernicious anemia, your doctor may change the type or dose of medicine you take. Infants of strict vegetarian mothers may be given vitamin B12 supplements from birth.

Sickle Cell Disease

The term sickle cell disease describes a group of inherited red blood cell disorders. People with sickle cell disease have abnormal hemoglobin, called hemoglobin S or sickle hemoglobin, in their red blood cells 4.

Hemoglobin is a protein in red blood cells that carries oxygen throughout the body.

“Inherited” means that the disease is passed by genes from parents to their children. Sickle Cell Disease is not contagious. A person cannot catch it, like a cold or infection, from someone else.

People who have sickle cell disease inherit two abnormal hemoglobin genes, one from each parent. In all forms of sickle cell disease, at least one of the two abnormal genes causes a person’s body to make hemoglobin S. When a person has two hemoglobin S genes, Hemoglobin SS, the disease is called sickle cell anemia. This is the most common and often most severe kind of sickle cell disease.

In the United States, most people with sickle cell disease are of African ancestry or identify themselves as black 5.

  • About 1 in 13 African American babies is born with sickle cell trait.
  • About 1 in every 365 black children is born with sickle cell disease.

There are also many people with this disease who come from Hispanic, southern European, Middle Eastern, or Asian Indian backgrounds.

Approximately 100,000 Americans have sickle cell disease.

Hemoglobin SC disease and hemoglobin Sβ thalassemia are two other common forms of sickle cell disease.

Some Forms of Sickle Cell Disease

  • Hemoglobin SS
  • Hemoglobin SC
  • Hemoglobin Sβ0 thalassemia
  • Hemoglobin Sβ+ thalassemia
  • Hemoglobin SD
  • Hemoglobin SE

Cells in tissues need a steady supply of oxygen to work well. Normally, hemoglobin in red blood cells takes up oxygen in the lungs and carries it to all the tissues of the body.

Red blood cells that contain normal hemoglobin are disc shaped (like a doughnut without a hole). This shape allows the cells to be flexible so that they can move through large and small blood vessels to deliver oxygen.

Sickle hemoglobin is not like normal hemoglobin. It can form stiff rods within the red cell, changing it into a crescent, or sickle shape.

Sickle-shaped cells are not flexible and can stick to vessel walls, causing a blockage that slows or stops the flow of blood. When this happens, oxygen can’t reach nearby tissues.

Figure 9. Sickle cell anemia

sickle cell anemia

The lack of tissue oxygen can cause attacks of sudden, severe pain, called pain crises. These pain attacks can occur without warning, and a person often needs to go to the hospital for effective treatment.

Most children with sickle cell disease are pain free between painful crises, but adolescents and adults may also suffer with chronic ongoing pain.

The red cell sickling and poor oxygen delivery can also cause organ damage. Over a lifetime, sickle cell disease can harm a person’s spleen, brain, eyes, lungs, liver, heart, kidneys, penis, joints, bones, or skin.

Sickle cells can’t change shape easily, so they tend to burst apart or hemolyze. Normal red blood cells live about 90 to 120 days, but sickle cells last only 10 to 20 days.

The body is always making new red blood cells to replace the old cells; however, in sickle cell disease the body may have trouble keeping up with how fast the cells are being destroyed. Because of this, the number of red blood cells is usually lower than normal. This condition, called anemia, can make a person have less energy.

Sickle cell disease is a life-long illness. The severity of the disease varies widely from person to person.

In high-income countries like the United States, the life expectancy of a person with sickle cell disease is now about 40–60 years. In 1973, the average lifespan of a person with sickle cell disease in the United States was only 14 years. Advances in the diagnosis and care of sickle cell disease have made this improvement possible.

At the present time, hematopoietic stem cell transplantation is the only cure for sickle cell disease 4. Unfortunately, most people with sickle cell disease are either too old for a transplant or don’t have a relative who is a good enough genetic match for them to act as a donor. A well-matched donor is needed to have the best chance for a successful transplant.

There are effective treatments that can reduce symptoms and prolong life. Early diagnosis and regular medical care to prevent complications also contribute to improved well-being.

What Causes Sickle Cell Disease ?

Abnormal hemoglobin, called hemoglobin S, causes sickle cell disease.

The problem in hemoglobin S is caused by a small defect in the gene that directs the production of the beta globin part of hemoglobin. This small defect in the beta globin gene causes a problem in the beta globin part of hemoglobin, changing the way that hemoglobin works.

How Is Sickle Cell Disease Inherited ?

When the hemoglobin S gene is inherited from only one parent and a normal hemoglobin gene is inherited from the other, a person will have sickle cell trait. People with sickle cell trait are generally healthy.

Only rarely do people with sickle cell trait have complications similar to those seen in people with sickle cell disease. But people with sickle cell trait are carriers of a defective hemoglobin S gene. So, they can pass it on when they have a child.

If the child’s other parent also has sickle cell trait or another abnormal hemoglobin gene (like thalassemia, hemoglobin C, hemoglobin D, hemoglobin E), that child has a chance of having sickle cell disease.

Figure 10. Sickle cell disease inheritance

sickle cell disease inheritance

[Source 6]

In the image above, each parent has one hemoglobin A gene and one hemoglobin S gene, and each of their children has:

  • A 25 percent chance of inheriting two normal genes: In this case the child does not have sickle cell trait or disease. (Blue)
  • A 50 percent chance of inheriting one hemoglobin A gene and one hemoglobin S gene: This child has sickle cell trait. (Purple)
  • A 25 percent chance of inheriting two hemoglobin S genes: This child has sickle cell disease. (Red)

It is important to keep in mind that each time this couple has a child, the chances of that child having sickle cell disease remain the same. In other words, if the first-born child has sickle cell disease, there is still a 25 percent chance that the second child will also have the disease. Both boys and girls can inherit sickle cell trait, sickle cell disease, or normal hemoglobin.

If a person wants to know if he or she carries a sickle hemoglobin gene, a doctor can order a blood test to find out.

What Are the Signs and Symptoms of Sickle Cell Disease ?

Early Signs and Symptoms

If a person has sickle cell disease, it is present at birth. But most infants do not have any problems from the disease until they are about 5 or 6 months of age. Every state in the United States, the District of Columbia, and the U.S. territories requires that all newborn babies receive screening for sickle cell disease. When a child has sickle cell disease, parents are notified before the child has symptoms.

Some children with sickle cell disease will start to have problems early on, and some later. Early symptoms of sickle cell disease may include:

  • Painful swelling of the hands and feet, known as dactylitis
  • Fatigue or fussiness from anemia
  • A yellowish color of the skin, known as jaundice, or whites of the eyes, known as icteris, that occurs when a large number of red cells hemolyze

The signs and symptoms of sickle cell disease will vary from person to person and can change over time. Most of the signs and symptoms of sickle cell disease are related to complications of the disease.

Major Complications of Sickle Cell Disease

Acute Pain (Sickle Cell or Vaso-occlusive) Crisis

Pain episodes (crises) can occur without warning when sickle cells block blood flow and decrease oxygen delivery. People describe this pain as sharp, intense, stabbing, or throbbing. Severe crises can be even more uncomfortable than post-surgical pain or childbirth.

Pain can strike almost anywhere in the body and in more than one spot at a time. But the pain often occurs in the

  • Lower back
  • Legs
  • Arms
  • Abdomen
  • Chest

A crisis can be brought on by

  • Illness
  • Temperature changes
  • Stress
  • Dehydration (not drinking enough)
  • Being at high altitudes

But often a person does not know what triggers, or causes, the crisis.

Chronic Pain

Many adolescents and adults with sickle cell disease suffer from chronic pain. This kind of pain has been hard for people to describe, but it is usually different from crisis pain or the pain that results from organ damage.

Chronic pain can be severe and can make life difficult. Its cause is not well understood.

Severe Anemia

People with sickle cell disease usually have mild to moderate anemia. At times, however, they can have severe anemia. Severe anemia can be life threatening. Severe anemia in an infant or child with sickle cell disease may be caused by:

  • Splenic sequestration crisis

The spleen is an organ that is located in the upper left side of the belly. The spleen filters germs in the blood, breaks up blood cells, and makes a kind of white blood cell. A splenic sequestration crisis occurs when red blood cells get stuck in the spleen, making it enlarge quickly. Since the red blood cells are trapped in the spleen, there are fewer cells to circulate in the blood. This causes severe anemia.

A big spleen may also cause pain in the left side of the belly. A parent can usually palpate or feel the enlarged spleen in the belly of his or her child.

  • Aplastic crisis

Aplastic crisis is usually caused by a parvovirus B19 infection, also called fifth disease or slapped cheek syndrome. Parvovirus B19 is a very common infection, but in sickle cell disease it can cause the bone marrow to stop producing new red cells for a while, leading to severe anemia.

Splenic sequestration crisis and aplastic crisis most commonly occur in infants and children with sickle cell disease. Adults with sickle cell disease may also experience episodes of severe anemia, but these usually have other causes.

No matter the cause, severe anemia may lead to symptoms that include:

  • Shortness of breath
  • Being very tired
  • Feeling dizzy
  • Having pale skin

Babies and infants with severe anemia may feed poorly and seem very sluggish.

  • Infections

The spleen is important for protection against certain kinds of germs. Sickle cells can damage the spleen and weaken or destroy its function early in life.

People with sickle cell disease who have damaged spleens are at risk for serious bacterial infections that can be life-threatening. Some of these bacteria include:

  • Pneumococcus
  • Hemophilus influenza type B
  • Meningococcus
  • Salmonella
  • Staphylococcus
  • Chlamydia
  • Mycoplasma pneumoniae

Bacteria can cause:

  • Blood infection (septicemia)
  • Lung infection (pneumonia)
  • Infection of the covering of the brain and spinal cord (meningitis)
  • Bone infection (osteomyelitis)

Acute Chest Syndrome

Sickling in blood vessels of the lungs can deprive a person’s lungs of oxygen. When this happens, areas of lung tissue are damaged and cannot exchange oxygen properly. This condition is known as acute chest syndrome. In acute chest syndrome, at least one segment of the lung is damaged.

This condition is very serious and should be treated right away at a hospital.

Acute chest syndrome often starts a few days after a painful crisis begins. A lung infection may accompany acute chest syndrome.

Symptoms may include:

  • Chest pain
  • Fever
  • Shortness of breath
  • Rapid breathing
  • Cough

Brain Complications

Clinical Stroke

A stroke occurs when blood flow is blocked to a part of the brain. When this happens, brain cells can be damaged or can die. In sickle cell disease, a clinical stroke means that a person shows outward signs that something is wrong. The symptoms depend upon what part of the brain is affected. Symptoms of stroke may include:

  • Weakness of an arm or leg on one side of the body
  • Trouble speaking, walking, or understanding
  • Loss of balance
  • Severe headache

As many as 24 percent of people with hemoglobin SS and 10 percent of people with hemoglobin SC may suffer a clinical stroke by age 45.

In children, clinical stroke occurs most commonly between the ages of 2 and 9, but recent prevention strategies have lowered the risk.

When people with sickle cell disease show symptoms of stroke, their families or friends should call your local emergency number right away.

Silent Stroke and Thinking Problems

Brain imaging and tests of thinking (cognitive studies) have shown that children and adults with hemoglobin SS and hemoglobin Sβ0 thalassemia often have signs of silent brain injury, also called silent stroke. Silent brain injury is damage to the brain without showing outward signs of stroke.

This injury is common. Silent brain injury can lead to learning problems or trouble making decisions or holding down a job.

Eye Problems

Sickle cell disease can injure blood vessels in the eye.

The most common site of damage is the retina, where blood vessels can overgrow, get blocked, or bleed. The retina is the light-sensitive layer of tissue that lines the inside of the eye and sends visual messages through the optic nerve to the brain.

Detachment of the retina can occur. When the retina detaches, it is lifted or pulled from its normal position. These problems can cause visual impairment or loss.

Heart Disease

People with sickle cell disease can have problems with blood vessels in the heart and with heart function. The heart can become enlarged. People can also develop pulmonary hypertension.

People with sickle cell disease who have received frequent blood transfusions may also have heart damage from iron overload.

Pulmonary Hypertension

In adolescents and adults, injury to blood vessels in the lungs can make it hard for the heart to pump blood through them. This causes the pressure in lung blood vessels to rise. High pressure in these blood vessels is called pulmonary hypertension. Symptoms may include shortness of breath and fatigue.

When this condition is severe, it has been associated with a higher risk of death.

Kidney Problems

The kidneys are sensitive to the effects of red blood cell sickling.

Sickle cell disease causes the kidneys to have trouble making the urine as concentrated as it should be. This may lead to a need to urinate often and to have bedwetting or uncontrolled urination during the night (nocturnal enuresis). This often starts in childhood. Other problems may include:

  • Blood in the urine
  • Decreased kidney function
  • Kidney disease
  • Protein loss in the urine

Priapism

Males with sickle cell disease can have unwanted, sometimes prolonged, painful erections. This condition is called priapism.

Priapism happens when blood flow out of the erect penis is blocked by sickled cells. If it goes on for a long period of time, priapism can cause permanent damage to the penis and lead to impotence.

If priapism lasts for more than 4 hours, emergency medical care should be sought to avoid complications.

Gallstones

When red cells hemolyze, they release hemoglobin. Hemoglobin gets broken down into a substance called bilirubin. Bilirubin can form stones that get stuck in the gallbladder. The gallbladder is a small, sac-shaped organ beneath the liver that helps with digestion. Gallstones are a common problem in sickle cell disease.

Gallstones may be formed early on but may not produce symptoms for years. When symptoms develop, they may include:

  • Right-sided upper belly pain
  • Nausea
  • Vomiting

If problems continue or recur, a person may need surgery to remove the gallbladder.

Liver Complications

There are a number of ways in which the liver may be injured in sickle cell disease.

Sickle cell intrahepatic cholestasis is an uncommon, but severe, form of liver damage that occurs when sickled red cells block blood vessels in the liver. This blockage prevents enough oxygen from reaching liver tissue.

These episodes are usually sudden and may recur. Children often recover, but some adults may have chronic problems that lead to liver failure.

People with sickle cell disease who have received frequent blood transfusions may develop liver damage from iron overload.

Leg Ulcers

Sickle cell ulcers are sores that usually start small and then get larger and larger.

The number of ulcers can vary from one to many. Some ulcers will heal quickly, but others may not heal and may last for long periods of time. Some ulcers come back after healing.

People with sickle cell disease usually don’t get ulcers until after the age of 10.

Joint Complications

Sickling in the bones of the hip and, less commonly, the shoulder joints, knees, and ankles, can decrease oxygen flow and result in severe damage. This damage is a condition called avascular or aseptic necrosis. This disease is usually found in adolescents and adults.

Symptoms include pain and problems with walking and joint movement. A person may need pain medicines, surgery, or joint replacement if symptoms persist.

Delayed Growth and Puberty

Children with sickle cell disease may grow and develop more slowly than their peers because of anemia. They will reach full sexual maturity, but this may be delayed.

Pregnancy

Pregnancies in women with sickle cell disease can be risky for both the mother and the baby.

Mothers may have medical complications including:

  • Infections
  • Blood clots
  • High blood pressure
  • Increased pain episodes

They are also at higher risk for:

  • Miscarriages
  • Premature births
  • “Small-for-dates babies” or underweight babies

Mental Health

As in other chronic diseases, people with sickle cell disease may feel sad and frustrated at times. The limitations that sickle cell disease can impose on a person’s daily activities may cause them to feel isolated from others. Sometimes they become depressed.

People with sickle cell disease may also have trouble coping with pain and fatigue, as well as with frequent medical visits and hospitalizations.

How Is Sickle Cell Disease Diagnosed ?

Screening Tests

People who do not know whether they make sickle hemoglobin (hemoglobin S) or another abnormal hemoglobin (such as C, β thalassemia, E) can find out by having their blood tested. This way, they can learn whether they carry a gene (i.e., have the trait) for an abnormal hemoglobin that they could pass on to a child.

When each parent has this information, he or she can be better informed about the chances of having a child with some type of sickle cell disease, such as hemoglobin SS, SC, Sβ thalassemia, or others.

Newborn Screening

When a child has sickle cell disease, it is very important to diagnose it early to better prevent complications.

Every state in the United States, the District of Columbia, and the U.S. territories require that every baby is tested for sickle cell disease as part of a newborn screening program.

In newborn screening programs, blood from a heel prick is collected in “spots” on a special paper. The hemoglobin from this blood is then analyzed in special labs.

Newborn screening results are sent to the doctor who ordered the test and to the child’s primary doctor.

If a baby is found to have sickle cell disease, health providers from a special follow-up newborn screening group contact the family directly to make sure that the parents know the results. The child is always retested to be sure that the diagnosis is correct.

Newborn screening programs also find out whether the baby has an abnormal hemoglobin trait. If so, parents are informed, and counseling is offered.

Remember that when a child has sickle cell trait or sickle cell disease, a future sibling, or the child’s own future child, may be at risk. These possibilities should be discussed with the primary care doctor, a blood specialist called a hematologist, and/or a genetics counselor.

Prenatal Screening

Doctors can also diagnose sickle cell disease before a baby is born. This is done using a sample of amniotic fluid, the liquid in the sac surrounding a growing embryo, or tissue taken from the placenta, the organ that attaches the umbilical cord to the mother’s womb.

Testing before birth can be done as early as 8–10 weeks into the pregnancy. This testing looks for the sickle hemoglobin gene rather than the abnormal hemoglobin.

How Is Sickle Cell Disease Treated ?

Health Maintenance To Prevent Complications

Babies with sickle cell disease should be referred to a doctor or provider group that has experience taking care of people with this disease. The doctor might be a hematologist (a doctor with special training in blood diseases) or an experienced general pediatrician, internist, or family practitioner.

For infants, the first sickle cell disease visit should take place before 8 weeks of age.

If someone was born in a country that doesn’t perform newborn sickle cell disease screening, he or she might be diagnosed with sickle cell disease later in childhood. These people should also be referred as soon as possible for special sickle cell disease care.

All people who have sickle cell disease should see their sickle cell disease care providers regularly. Regularly means every 3 to 12 months, depending on the person’s age. The sickle cell disease doctor or team can help to prevent problems by:

  • Examining the person
  • Giving medicines and immunizations
  • Performing tests
  • Educating families about the disease and what to watch out for

Preventing Infection

In sickle cell disease, the spleen doesn’t work properly or doesn’t work at all. This problem makes people with sickle cell disease more likely to get severe infections.

Penicillin

In children with sickle cell disease, taking penicillin two times a day has been shown to reduce the chance of having a severe infection caused by the pneumococcus bacteria. Infants need to take liquid penicillin. Older children can take tablets.

Many doctors will stop prescribing penicillin after a child has reached the age of 5. Some prefer to continue this antibiotic throughout life, particularly if a person has hemoglobin SS or hemoglobin Sβ0 thalassemia, since people with sickle cell disease are still at risk. All people who have had surgical removal of the spleen, called a splenectomy, or a past infection with pneumococcus should keep taking penicillin throughout life.

Immunizations

People with sickle cell diseaseshould receive all recommended childhood vaccines. They should also receive additional vaccines to prevent other infections.

Pneumococcus. Even though all children routinely receive the vaccine against pneumococcus (PCV13), children with sickle cell disease should also receive a second kind of vaccine against pneumococcus (PPSV23). This second vaccine is given after 24 months of age and again 5 years later. Adults with sickle cell disease who have not received any pneumococcal vaccine should get a dose of the PCV13 vaccine. They should later receive the PPSV23 if they have not already received it or it has been more than 5 years since they did. A person should follow these guidelines even if he or she is still taking penicillin.

Influenza. All people with sickle cell disease should receive an influenza shot every year at the start of flu season. This should begin at 6 months of age. Only the inactivated vaccine, which comes as a shot, should be used in people with sickle cell disease.

Meningococcus. A child with sickle cell disease should receive this vaccine (Menactra or Menveo) at 2, 4, 6, and 12–15 months of age. The child should receive a booster vaccine 3 years after this series of shots, then every 5 years after that.

Screening Tests and Evaluations

Height, Weight, Blood Pressure, and Oxygen Saturation

Doctors will monitor height and weight to be sure that a child is growing properly and that a person with sickle cell disease is maintaining a healthy weight.

Doctors will also track a person’s blood pressure. When a person with sickle cell disease has high blood pressure, it needs to be treated promptly because it can increase the risk of stroke.

Oxygen saturation testing provides information about how much oxygen the blood is carrying.

Blood and Urine Testing

People with sickle cell disease need to have frequent lab tests.

Blood tests help to establish a person’s “baseline” for problems like anemia. Blood testing also helps to show whether a person has organ damage, so that it can be treated early.

Urine testing can help to detect early kidney problems or infections.

Transcranial Doppler Ultrasound Screening

Children who have hemoglobin SS or hemoglobin Sβ0 thalassemia and are between the ages of 2 and 16 should have Transcranial Doppler Ultrasound testing once a year.

This study can find out whether a child is at higher risk for stroke. When the test is abnormal, regular blood transfusions can decrease the chances of having a stroke.

The child is awake during the Transcranial Doppler Ultrasound exam. The test does not hurt at all. The Transcranial Doppler Ultrasound machine uses sound waves to measure blood flow like the ultrasound machine used to examine pregnant women.

Eye Examinations

An eye doctor, or ophthalmologist, should examine a person’s eyes every 1—2 years from the age of 10 onwards.

These exams can detect if there are sickle cell disease-related problems of the eye. Regular exams can help doctors find and treat problems early to prevent loss of vision. A person should see his or her doctor right away for any sudden change in vision.

Pulmonary Hypertension

Doctors have different approaches to screening for pulmonary hypertension. This is because studies have not given clear information as to when and how a person should receive the screening. People with sickle cell disease and their caretakers should discuss with their doctor whether screening makes sense for them.

Cognitive Screening

People with sickle cell disease can develop cognitive (thinking) problems that may be hard to notice early in life.

Sometimes these problems are caused by “silent” strokes that can only be seen with magnetic resonance imaging (MRI) of the brain.

People with sickle cell disease should tell their doctors or nurses if they have thinking problems, such as difficulties learning in school, slowed decision making, or trouble organizing their thoughts.

People can be referred for cognitive testing. This testing can identify areas in which a person could use extra help.

Children with sickle cell disease who have thinking problems may qualify for an Individualized Education Program. An Individualized Education Program is a plan that helps students to reach their educational goals. Adults may be able to enroll in vocational rehabilitation programs that can help them with job training.

Education and Guidance

Doctors and other providers will talk with people who have sickle cell disease and their caretakers about complications and also review information at every visit.

Because there are a lot of things to discuss, new topics are often introduced as a child or adult reaches an age when that subject is important to know about.

Doctors and nurses know that there is a lot of information to learn, and they don’t expect people to know everything after one discussion. People with sickle cell disease and their families should not be afraid to ask questions.

Topics that are usually covered include:

  • Hours that medical staff are available and contact information to use when people with sickle cell disease or caretakers have questions
  • A plan for what to do and where to get care if a person has a fever, pain, or other signs of sickle cell disease complications that need immediate attention
  • How sickle cell disease is inherited and the risk of having a child with sickle cell disease
  • The importance of regular medical visits, screening tests, and evaluations
  • How to recognize and manage pain
  • How to palpate (feel) a child’s spleen. Because of the risk of splenic sequestration crisis, caretakers should learn how to palpate a child’s spleen. They should try to feel for the spleen daily and more frequently when the child is ill. If they feel that the spleen is bigger than usual, they should call the care provider.

Transitioning Care

When children with sickle cell disease become adolescents or young adults, they often need to transition from a pediatric care team to an adult care team. This period has been shown to be associated with increased hospital admissions and medical problems. There seem to be many reasons for this.

Some of the increased risk is directly related to the disease. As people with sickle cell disease get older, they often develop more organ damage and more disabilities.

The shift in care usually occurs at the same time that adolescents are undergoing many changes in their emotional, social, and academic lives. The transition to more independent self-management may be difficult, and following treatment plans may become less likely.

When compared with pediatrics, there are often fewer adult sickle cell disease programs available in a given region. This makes it more difficult for a person with sickle cell disease to find appropriate doctors, particularly those with whom they feel comfortable.

To improve use of regular medical care by people with sickle cell disease and to reduce age-related complications, many sickle cell disease teams have developed special programs that the make transition easier. Such programs should involve the pediatric and the adult care teams. They should also start early and continue over several years.

Managing Some Complications of sickle cell disease

Acute Pain

Each person with sickle cell disease should have a home treatment regimen that is best suited to their needs. The providers on the sickle cell disease team usually help a person develop a written, tailored care plan. If possible, the person with sickle cell disease should carry this plan with them when they go to the emergency room.

When an acute crisis is just starting, most doctors will advise the person to drink lots of fluids and to take a non-steroidal anti-inflammatory (NSAID) pain medication, such as ibuprofen. When a person has kidney problems, acetaminophen is often preferred.

If pain persists, many people will find that they need a stronger medicine.

Combining additional interventions, such as massage, relaxation methods, or a heating pad, may also help.

If a person with sickle cell disease cannot control the pain at home, he or she should go to an sickle cell disease day hospital/outpatient unit or an emergency room to receive additional, stronger medicines and intravenous (IV) fluids.

Some people may be able to return home once their pain is under better control. In this case, the doctor may prescribe additional pain medicines for a short course of therapy.

People often need to be admitted to the hospital to fully control an acute pain crisis.

When taken daily, hydroxyurea has been found to decrease the number and severity of pain episodes.

Some patients may have fewer visits to the hospital or hospitalizations due to severe pain, and may have shorter hospital stays for pain crises if they are taking L-glutamine oral powder (Endari) compared to patients who are not taking this medicine. More research is needed to understand how effective L-glutamine oral powder is as a treatment and which patients may benefit from using it.

Chronic Pain

Sometimes chronic pain results from a complication, such as a leg ulcer or aseptic necrosis of the hip. In this case, doctors try to treat the complication causing the pain.

While chronic pain is common in adults with sickle cell disease, the cause is often poorly understood. Taking pain medicines daily may help to decrease the pain. Some examples of these medicines include:

  • NSAID drugs, such as ibuprofen
  • Duloxetine
  • Gabapentin
  • Amitriptyline
  • Strong pain medicines, such as opiates

Other approaches, such as massage, heat, or acupuncture may be helpful in some cases. Chronic pain often comes with feelings of depression and anxiety. Supportive counseling and, sometimes, antidepressant medicines may help.

Severe Anemia

People should see their doctors or go to a hospital right away if they develop anemia symptoms from a splenic sequestration crisis or an aplastic crisis. These conditions can be life-threatening, and the person will need careful monitoring and treatment in the hospital. A person also usually needs a blood transfusion.

People with sickle cell disease and symptoms of severe anemia from other causes should also see a doctor right away.

Some patients may have fewer hospital visits due to sickle cell crises, including splenic sequestration, if they are taking L-glutamine oral powder compared to patients who are not taking this medicine. More research is needed to understand how effective L-glutamine oral powder is as a treatment and which patients may benefit from using it.

Infections

Fever is a medical emergency in sickle cell disease. All caretakers of infants and children with sickle cell disease should take their child to their doctor or go to a hospital right away when their child has a fever. Adults with sickle cell disease should also seek care for fever or other signs of infection.

All children and adults who have sickle cell disease and a fever (over 38.50 C or 101.30 F) must be seen by a doctor and treated with antibiotics right away.

Some people will need to be hospitalized, while others may receive care and follow-up as an outpatient.

Acute Chest Syndrome

People with sickle cell disease and symptoms of acute chest syndrome should see their doctor or go to a hospital right away.

They will need to be admitted to the hospital where they should receive antibiotics and close monitoring. They may need oxygen therapy and a blood transfusion.

When taken daily, the medicine hydroxyurea has been found to decrease the number and severity of acute chest events.

Some patients may have fewer hospital visits due to sickle cell crises, including acute chest syndrome, if they are taking L-glutamine oral powder compared to patients who are not taking this medicine. More research is needed to understand how effective L-glutamine oral powder is as a treatment and which patients may benefit from using it.

Clinical Stroke

People with sickle cell disease who have symptoms of stroke should be brought to the hospital right away by an ambulance. If a person is having symptoms of stroke, someone should call your local emergency number.

Symptoms of stroke may include:

  • Weakness of an arm or leg on one side of the body
  • Trouble speaking, walking, or understanding
  • Loss of balance
  • Severe headache

If imaging studies reveal that the person has had an acute stroke, he or she may need an exchange transfusion. This procedure involves slowly removing an amount of the person’s blood and replacing it with blood from a donor who does not have sickle cell disease or sickle cell trait. Afterward, the person may need to receive monthly transfusions or other treatments to help to prevent another stroke.

Silent Stroke and Cognitive Problems

Children and adults with sickle cell disease and cognitive problems may be able to get useful help based upon the results of their testing. For instance, children may qualify for an Individualized Education Program. Adults may be able to enroll in vocational, or job, training programs.

Priapism

Sometimes, a person may be able to relieve priapism by:

  • Emptying the bladder by urinating
  • Taking medicine
  • Increasing fluid intake
  • Doing light exercise

If a person has an episode that lasts for 4 hours or more, he should go to the hospital to see a hematologist and urologist.

Some patients may have fewer hospital visits due to sickle cell crises, including priapism, if they are taking L-glutamine oral powder compared to patients who are not taking this medicine. More research is needed to understand how effective L-glutamine oral powder is as a treatment and which patients may benefit from using it.

Pregnancy

Pregnant women with sickle cell disease are at greater risk for problems. They should always see an obstetrician who has experience with sickle cell disease and high-risk pregnancies and deliveries.

The obstetrician should work with a hematologist or primary medical doctor who is well informed about sickle cell disease and its complications.

Pregnant women with sickle cell disease need more frequent medical visits so that their doctors can follow them closely. The doctor may prescribe certain vitamins and will be careful to prescribe pain medicines that are safe for the baby.

A pregnant woman with sickle cell disease may need to have one or more blood transfusions during her pregnancy to treat complications, such as worsening anemia or an increased number of pain or acute chest syndrome events.

Hydroxyurea

Hydroxyurea is an oral medicine that has been shown to reduce or prevent several sickle cell disease complications.

This medicine was studied in patients with sickle cell disease because it was known to increase the amount of fetal hemoglobin (hemoglobin F) in the blood. Increased hemoglobin F provides some protection against the effects of hemoglobin S.

Hydroxyurea was later found to have several other benefits for a person with sickle cell disease, such as decreasing inflammation.

Use in adults. Many studies of adults with hemoglobin SS or hemoglobin Sβ thalassemia showed that hydroxyurea reduced the number of episodes of pain crises and acute chest syndrome. It also improved anemia and decreased the need for transfusions and hospital admissions.
Use in children. Studies in children with severe hemoglobin SS or Sβ thalassemia showed that hydroxyurea reduced the number of vaso-occlusive crises and hospitalizations. A study of very young children (between the ages of 9 and 18 months) with hemoglobin SS or hemoglobin Sβ thalassemia also showed that hydroxyurea decreased the number of episodes of pain and dactylitis.

Who Should Use Hydroxyurea ?

Since hydroxyurea can decrease several complications of sickle cell disease, most experts recommend that children and adults with hemoglobin SS or Sβ0 thalassemia who have frequent painful episodes, recurrent chest crises, or severe anemia take hydroxyurea daily.

Some experts offer hydroxyurea to all infants over 9 months of age and young children with hemoglobin SS or Sβ0 thalassemia, even if they do not have severe clinical problems, to prevent or reduce the chance of complications. There is no information about how safe or effective hydroxyurea is in children under 9 months of age.

Some experts will prescribe hydroxyurea to people with other types of sickle cell disease who have severe, recurrent pain. There is little information available about how effective hydroxyurea is for these types of sickle cell disease.

In all situations, people with sickle cell disease should discuss with their doctors whether or not hydroxyurea is an appropriate medication for them.

Pregnant women should not use hydroxyurea.

How Is Hydroxyurea Taken ?

To work properly, hydroxyurea should be taken by mouth daily at the prescribed dose. When a person does not take it regularly, it will not work as well, or it won’t work at all.

A person with sickle cell disease who is taking hydroxyurea needs careful monitoring. This is particularly true in the early weeks of taking the medicine. Monitoring includes regular blood testing and dose adjustments.

What Are the Risks of Hydroxyurea ?

Hydroxyurea can cause the blood’s white cell count or platelet count to drop. In rare cases, it can worsen anemia. These side effects usually go away quickly if a person stops taking the medication. When a person restarts it, a doctor usually prescribes a lower dose.

Other short-term side effects are less common.

It is still unclear whether hydroxyurea can cause problems later in life in people with sickle cell disease who take it for many years. Studies so far suggest that it does not put people at a higher risk of cancer and does not affect growth in children. But further studies are needed.

Red Blood Cell Transfusions

Doctors may use acute and chronic red blood cell transfusions to treat and prevent certain sickle cell disease complications. The red blood cells in a transfusion have normal hemoglobin in them.

A transfusion helps to raise the number of red blood cells and provides normal red blood cells that are more flexible than red blood cells with sickle hemoglobin. These cells live longer in the circulation. Red blood cell transfusions decrease vaso-occlusion (blockage in the blood vessel) and improve oxygen delivery to the tissues and organs.

Acute Transfusion in sickle cell disease

Doctors use blood transfusions in sickle cell disease for complications that cause severe anemia. They may also use them when a person has an acute stroke, in many cases of acute chest crises, and in multi-organ failure.

A person with sickle cell disease usually receives blood transfusions before surgery to prevent sickle cell disease-related complications afterwards.

Chronic Transfusion

Doctors recommend regular or ongoing blood transfusions for people who have had an acute stroke, since transfusions decrease the chances of having another stroke.

Doctors also recommend chronic blood transfusions for children who have abnormal Transcranial Doppler Ultrasound results because transfusions can reduce the chance of having a first stroke.

Some doctors use this approach to treat complications that do not improve with hydroxyurea. They may also use transfusions in people who have too many side effects from hydroxyurea.

What Are the Risks of Transfusion Therapy ?

Possible complications include:

  • Hemolysis
  • Iron overload, particularly in people receiving chronic transfusions (can severely impair heart and lung function)
  • Infection
  • Alloimmunization (can make it hard to find a matching unit of blood for a future transfusion)

All blood banks and hospital personnel have adopted practices to reduce the risk of transfusion problems.

People with sickle cell disease who receive transfusions should be monitored for and immunized against hepatitis. They should also receive regular screenings for iron overload. If a person has iron overload, the doctor will give chelation therapy, a medicine to reduce the amount of iron in the body and the problems that iron overload causes.

Hematopoietic Stem Cell Transplantation

At the present time, hematopoietic stem cell transplantation (HSCT) is the only cure for sickle cell disease. People with sickle cell disease and their families should ask their doctor about this procedure.

What Are Stem Cells ?

Stem cells are special cells that can divide over and over again. After they divide, these cells can go on to become blood red cells, white cells, or platelets.

A person with sickle cell disease has stem cells that make red blood cells that can sickle. People without sickle cell disease have stem cells that make red cells that usually won’t sickle.

What Stem Cells Are Used in hematopoietic stem cell transplantation ?

In hematopoietic stem cell transplantation, stem cells are taken from the bone marrow or blood of a person who does not have sickle cell disease (the donor). The donor, however, may have sickle cell trait.

The donor is often the person’s sister or brother. This is because the safest and most successful transplants use stem cells that are matched for special proteins called HLA antigens. Since these antigens are inherited from parents, a sister or brother is the most likely person to have the same antigens as the person with sickle cell disease.

What Happens During hematopoietic stem cell transplantation ?

First, stem cells are taken from the donor. After this, the person with sickle cell disease (the recipient) is treated with drugs that destroy or reduce his or her own bone marrow stem cells.

The donor stem cells are then injected into the person’s vein. The injected cells will make a home in the recipient’s bone marrow, gradually replacing the recipient’s cells. The new stem cells will make red cells that do not sickle.

Which People Receive hematopoietic stem cell transplantation ?

At the present time, most sickle cell disease transplants are performed in children who have had complications such as strokes, acute chest crises, and recurring pain crises. These transplants usually use a matched donor.

Because only about 1 in 10 children with sickle cell disease has a matched donor without sickle cell disease in their families, the number of people with sickle cell disease who get transplants is low.

Hematopoietic stem cell transplantation is more risky in adults, and that is why most transplants are done in children.

There are several medical centers that are researching new sickle cell disease hematopoietic stem cell transplantation techniques in children and adults who don’t have a matched donor in the family or are older than most recipients. Hopefully, more people with sickle cell disease will be able to receive a transplant in the future, using these new methods.

What Are the Risks ?

Hematopoietic stem cell transplantation is successful in about 85 percent of children when the donor is related and HLA matched. Even with this high success rate, hematopoietic stem cell transplantation still has risks.

Complications can include severe infections, seizures, and other clinical problems. About 5 percent of people have died. Sometimes transplanted cells attack the recipient’s organs (graft versus host disease).

Medicines are given to prevent many of the complications, but they still can happen.

What happens when you have too many red blood cells

Polycythemia vera is a rare blood disease in which your your bone marrow makes too many red blood cells and it also can make too many white blood cells and platelets 7. The disease affects people of all ages, but it’s most common in adults who are older than 60. Polycythemia vera is rare in children and young adults. Men are at slightly higher risk for polycythemia vera than women.

The extra red blood cells make your blood thicker than normal. As a result, blood clots can form more easily. These clots can block blood flow through your arteries and veins, which can cause a heart attack or stroke.

Thicker blood also doesn’t flow as quickly to your body as normal blood. Slowed blood flow prevents your organs from getting enough oxygen, which can cause serious problems, such as angina (heart pain) and heart failure. Angina is chest pain or discomfort.

A mutation, or change, in the body’s JAK2 gene is the major cause of polycythemia vera. This gene makes a protein that helps the body produce blood cells. What causes the change in the JAK2 gene isn’t known. Polycythemia vera generally isn’t inherited—that is, passed from parents to children through genes.

Polycythemia vera develops slowly and may not cause symptoms for years. The disease often is found during routine blood tests done for other reasons.

When signs and symptoms are present, they’re the result of the thick blood that occurs with polycythemia vera. This thickness slows the flow of oxygen-rich blood to all parts of your body. Without enough oxygen, many parts of your body won’t work normally.

For example, slower blood flow deprives your arms, legs, lungs, and eyes of the oxygen they need. This can cause headaches, dizziness, itching, and vision problems, such as blurred or double vision.

Polycythemia vera is a serious, chronic (ongoing) disease that can be fatal if not diagnosed and treated. Polycythemia vera has no cure, but treatments can help control the disease and its complications.

Polycythemia vera is treated with procedures, medicines, and other methods. You may need one or more treatments to manage the disease.

What Causes Polycythemia Vera ?

Primary Polycythemia

Polycythemia vera also is known as primary polycythemia. A mutation, or change, in the body’s JAK2 gene is the main cause of polycythemia vera. The JAK2 gene makes a protein that helps the body produce blood cells.

What causes the change in the JAK2 gene isn’t known. Polycythemia vera generally isn’t inherited—that is, passed from parents to children through genes. However, in some families, the JAK2 gene may have a tendency to mutate. Other, unknown genetic factors also may play a role in causing polycythemia vera.

Secondary Polycythemia

Another type of polycythemia, called secondary polycythemia, isn’t related to the JAK2 gene. Long-term exposure to low oxygen levels causes secondary polycythemia.

A lack of oxygen over a long period can cause your body to make more of the hormone erythropoietin (EPO). High levels of EPO can prompt your body to make more red blood cells than normal. This leads to thicker blood, as seen in polycythemia vera.

People who have severe heart or lung disease may develop secondary polycythemia. People who smoke, spend long hours at high altitudes, or are exposed to high levels of carbon monoxide where they work or live also are at risk.

For example, working in an underground parking garage or living in a home with a poorly vented fireplace or furnace can raise your risk for secondary polycythemia.

Rarely, tumors can make and release EPO, or certain blood problems can cause the body to make more EPO.

Sometimes doctors can cure secondary polycythemia—it depends on whether the underlying cause can be stopped, controlled, or cured.

What Are the Signs and Symptoms of Polycythemia Vera ?

Polycythemia vera develops slowly. The disease may not cause signs or symptoms for years.

When signs and symptoms are present, they’re the result of the thick blood that occurs with polycythemia vera. This thickness slows the flow of oxygen-rich blood to all parts of your body. Without enough oxygen, many parts of your body won’t work normally.

The signs and symptoms of polycythemia vera include:

  • Headaches, dizziness, and weakness
  • Shortness of breath and problems breathing while lying down
  • Feelings of pressure or fullness on the left side of the abdomen due to an enlarged spleen (an organ in the abdomen)
  • Double or blurred vision and blind spots
  • Itching all over (especially after a warm bath), reddened face, and a burning feeling on your skin (especially your hands and feet)
  • Bleeding from your gums and heavy bleeding from small cuts
  • Unexplained weight loss
  • Fatigue (tiredness)
  • Excessive sweating
  • Very painful swelling in a single joint, usually the big toe (called gouty arthritis)

In rare cases, people who have polycythemia vera may have pain in their bones.

Polycythemia Vera Complications

If you have polycythemia vera, the thickness of your blood and the slowed blood flow can cause serious health problems.

Blood clots are the most serious complication of polycythemia vera. Blood clots can cause a heart attack or stroke. They also can cause your liver and spleen to enlarge. Blood clots in the liver and spleen can cause sudden, intense pain.

Slowed blood flow also prevents enough oxygen-rich blood from reaching your organs. This can lead to angina (chest pain or discomfort) and heart failure. The high levels of red blood cells that polycythemia vera causes can lead to stomach ulcers, gout, or kidney stones.

Some people who have polycythemia vera may develop myelofibrosis. This is a condition in which your bone marrow is replaced with scar tissue. Abnormal bone marrow cells may begin to grow out of control.

This abnormal growth can lead to acute myelogenous leukemia (AML), a cancer of the blood and bone marrow. This disease can worsen very quickly.

How Is Polycythemia Vera Diagnosed ?

Polycythemia vera may not cause signs or symptoms for years. The disease often is found during routine blood tests done for other reasons. If the results of your blood tests aren’t normal, your doctor may want to do more tests.

Your doctor will diagnose polycythemia vera based on your signs and symptoms, your age and overall health, your medical history, a physical exam, and test results.

During the physical exam, your doctor will look for signs of polycythemia vera. He or she will check for an enlarged spleen, red skin on your face, and bleeding from your gums.

If your doctor confirms that you have polycythemia, the next step is to find out whether you have primary polycythemia or secondary polycythemia.

Your medical history and physical exam may confirm which type of polycythemia you have. If not, you may have tests that check the level of the hormone erythropoietin (EPO) in your blood.

People who have polycythemia vera have very low levels of EPO. People who have secondary polycythemia usually have normal or high levels of EPO.

Specialists Involved

If your primary care doctor thinks you have polycythemia vera, he or she may refer you to a hematologist. A hematologist is a doctor who specializes in diagnosing and treating blood diseases and conditions.

Diagnostic Tests

You may have blood tests to diagnose polycythemia vera. These tests include a complete blood count (CBC) and other tests, if necessary.
Complete Blood Count

Often, the first test used to diagnose polycythemia vera is a CBC. The CBC measures many parts of your blood.

This test checks your hemoglobin and hematocrit levels. Hemoglobin is an iron-rich protein that helps red blood cells carry oxygen from the lungs to the rest of the body. Hematocrit is a measure of how much space red blood cells take up in your blood. A high level of hemoglobin or hematocrit may be a sign of polycythemia vera.

The CBC also checks the number of red blood cells, white blood cells, and platelets in your blood. Abnormal results may be a sign of polycythemia vera, a blood disorder, an infection, or another condition.

In addition to high red blood cell counts, people who have polycythemia vera also may have high white blood cell and/or platelet counts.

Other Blood Tests

Blood smear. For this test, a small sample of blood is drawn from a vein, usually in your arm. The blood sample is examined under a microscope.

A blood smear can show whether you have a higher than normal number of red blood cells. The test also can show abnormal blood cells that are linked to myelofibrosis and other conditions related to polycythemia vera.

Erythropoietin level. This blood test measures the level of EPO in your blood. EPO is a hormone that prompts your bone marrow to make new blood cells. People who have polycythemia vera have very low levels of EPO. People who have secondary polycythemia usually have normal or high levels of EPO.

Bone Marrow Tests

Bone marrow tests can show whether your bone marrow is healthy. These tests also show whether your bone marrow is making normal amounts of blood cells.

The two bone marrow tests are aspiration and biopsy. For aspiration, your doctor removes a small amount of fluid bone marrow through a needle. For a biopsy, your doctor removes a small amount of bone marrow tissue through a larger needle. The samples are then examined under a microscope.

If the tests show that your bone marrow is making too many blood cells, it may be a sign that you have polycythemia vera.

How Is Polycythemia Vera Treated ?

Polycythemia vera doesn’t have a cure. However, treatments can help control the disease and its complications. polycythemia vera is treated with procedures, medicines, and other methods. You may need one or more treatments to manage the disease.

Goals of Treatment

The goals of treating polycythemia vera are to control symptoms and reduce the risk of complications, especially heart attack and stroke. To do this, polycythemia vera treatments reduce the number of red blood cells and the level of hemoglobin (an iron-rich protein) in the blood. This brings the thickness of your blood closer to normal.

Blood with normal thickness flows better through the blood vessels. This reduces the chance that blood clots will form and cause a heart attack or stroke.

Blood with normal thickness also ensures that your body gets enough oxygen. This can help reduce some of the signs and symptoms of polycythemia vera, such as headaches, vision problems, and itching.

Studies show that treating polycythemia vera greatly improves your chances of living longer.

The goal of treating secondary polycythemia is to control its underlying cause, if possible. For example, if the cause is carbon monoxide exposure, the goal is to find the source of the carbon monoxide and fix or remove it.

Treatments To Lower Red Blood Cell Levels

Phlebotomy

Phlebotomy is a procedure that removes some blood from your body. For this procedure, a needle is inserted into one of your veins. Blood from the vein flows through an airtight tube into a sterile container or bag. The process is similar to the process of donating blood.

Phlebotomy reduces your red blood cell count and starts to bring your blood thickness closer to normal.

Typically, a pint (1 unit) of blood is removed each week until your hematocrit level approaches normal. Hematocrit is the measure of how much space red blood cells take up in your blood.

You may need to have phlebotomy done every few months.

Medicines

Your doctor may prescribe medicines to keep your bone marrow from making too many red blood cells. Examples of these medicines include hydroxyurea and interferon-alpha.

Hydroxyurea is a medicine generally used to treat cancer. This medicine can reduce the number of red blood cells and platelets in your blood. As a result, this medicine helps improve your blood flow and bring the thickness of your blood closer to normal.

Interferon-alpha is a substance that your body normally makes. It also can be used to treat polycythemia vera. Interferon-alpha can prompt your immune system to fight overactive bone marrow cells. This helps lower your red blood cell count and keep your blood flow and blood thickness closer to normal.

Radiation Treatment

Radiation treatment can help suppress overactive bone marrow cells. This helps lower your red blood cell count and keep your blood flow and blood thickness closer to normal.

However, radiation treatment can raise your risk of leukemia (blood cancer) and other blood diseases.

Treatments for Symptoms

Aspirin can relieve bone pain and burning feelings in your hands or feet that you may have as a result of polycythemia vera. Aspirin also thins your blood, so it reduces the risk of blood clots.

Aspirin can have side effects, including bleeding in the stomach and intestines. For this reason, take aspirin only as your doctor recommends.

If your polycythemia vera causes itching, your doctor may prescribe medicines to ease the discomfort. Your doctor also may prescribe ultraviolet light treatment to help relieve your itching.

Other ways to reduce itching include:

  • Avoiding hot baths. Cooler water can limit irritation to your skin.
  • Gently patting yourself dry after bathing. Vigorous rubbing with a towel can irritate your skin.
  • Taking starch baths. Add half a box of starch to a tub of lukewarm water. This can help soothe your skin.

Experimental Treatments

Researchers are studying other treatments for polycythemia vera. An experimental treatment for itching involves taking low doses of selective serotonin reuptake inhibitors (SSRIs). This type of medicine is used to treat depression. In clinical trials, SSRIs reduced itching in people who had polycythemia vera.

Imatinib mesylate is a medicine that’s approved for treating leukemia. In clinical trials, this medicine helped reduce the need for phlebotomy in people who had polycythemia vera. This medicine also helped reduce the size of enlarged spleens.

Researchers also are trying to find a treatment that can block or limit the effects of an abnormal JAK2 gene. A mutation, or change, in the JAK2 gene is the major cause of polycythemia vera.

Blood Transfusions

Early attempts to transfer blood from one person to another produced varied results. Sometimes, the recipient improved. Other times, the recipient suffered a blood transfusion reaction in which the red blood cells clumped, obstructing vessels and producing great pain and organ damage.

Eventually scientists determined that blood is of differing types and that only certain combinations of blood types are compatible. These discoveries led to the development of procedures for typing blood. Today, safe transfusions of whole blood depend on two blood tests—“type and cross match.” First the recipient’s ABO blood type and Rh status are determined. Following this, a “cross match” is done of the recipient’s serum with a small amount of the donor’s red blood cells that have the same ABO type and Rh status as the recipient. Compatibility is determined by examining the mixture under a microscope for agglutination, the clumping of red blood cells. Agglutination (“clumping”) is a reaction between antigens and specific antibodies. This is what happens when antigens on mismatched donated red blood cells react with antibodies in plasma.

Blood Antigens and Antibodies

An antigen, is any molecule that triggers an immune response, the body’s reaction to invasion by a foreign substance or organism. When the immune system encounters an antigen not found on the body’s own cells, it will attack, producing antibodies. In a transfusion reaction, antigens (agglutinogens) on the surface of the donated red blood cells react with antibodies (agglutinins) in the plasma of the recipient, resulting in the agglutination of the donated red blood cells.

A mismatched blood transfusion quickly produces telltale signs of agglutination—anxiety, breathing difficulty, facial flushing, headache, and severe pain in the neck, chest, and lumbar area. Red blood cells burst, releasing free hemoglobin. Liver cells and macrophages phagocytize the hemoglobin, converting it to bilirubin, which may sufficiently accumulate to cause the yellow skin of jaundice. Free hemoglobin reaching the kidneys may ultimately cause them to fail.

Only a few of the 32 known antigens on red blood cell membranes can produce serious transfusion reactions. These include the antigens of the ABO group and those of the Rh group. Avoiding the mixture of certain kinds of antigens and antibodies prevents adverse transfusion reactions.

ABO Blood Group

The ABO blood group is based on the presence (or absence) of two major antigens on red blood cell membranes—antigen A and antigen B. A and B antigens are carbohydrates attached to glycolipids projecting from the red blood cell surface. A person’s erythrocytes have on their surfaces one of four antigen combinations: only A, only B, both A and B, or neither A nor B.

A person with only antigen A has type A blood. A person with only antigen B has type B blood. An individual with both antigens A and B has type AB blood. A person with neither antigen A nor B has type O blood. Thus, all people have one of four possible ABO blood types—A, B, AB, or O. The resulting ABO blood type is inherited. It is the consequence of DNA encoding enzymes to synthesize A or B antigens on erythrocytes in one of these four combinations.

  • In the United States, the most common ABO blood types are O (47%) and A (41%). Rarer are type B (9%) and type AB (3%). These percentages vary in subpopulations and over time, reflecting changes in the genetic structure of populations.

Antibodies that affect the ABO blood group antigens are present in the plasma about two to eight months following birth, as a result of exposure to foods or microorganisms containing the antigen(s) that are not present on the individual’s red blood cells. Specifically, whenever antigen A is absent in red blood cells, an antibody called anti-A is produced, and whenever antigen B is absent on cells, an antibody called anti-B is produced. Therefore, individuals with type A blood have anti-B antibody in their plasma; those with type B blood have anti-A antibody; those with type AB blood have neither antibody; and those with type O blood have both anti-A and anti-B antibodies (Figure 11 and Table 1). The antibodies anti-A and anti-B are large and do not cross the placenta. Thus, a pregnant woman and her fetus may be of different ABO blood types, but agglutination in the fetus will not occur.

An antibody of one type will react with an antigen of the same type and clump red blood cells; therefore, such combinations must be avoided. The major concern in blood transfusion procedures is that the cells in the donated blood not clump due to antibodies in the recipient’s plasma. For this reason, a person with type A (anti-B) blood must not receive blood of type B or AB, either of which would clump in the presence of anti-B in the recipient’s type A blood. Likewise, a person with type B (anti-A) blood must not receive type A or AB blood, and a person with type O (anti-A and anti-B) blood must not receive type A, B, or AB blood.

Type AB blood does not have anti-A or anti-B antibodies, so an AB person can receive a transfusion of blood of any other type. For this reason, individuals with type AB blood are called universal recipients. However, type A (anti-B) blood, type B (anti-A) blood, and type O (anti-A and anti-B) blood still contain antibodies (either anti-A and/or anti-B) that could agglutinate type AB cells if transfused rapidly.

Consequently, even for AB individuals, using donor blood of the same type as the recipient is best. Type O blood has neither antigen A nor antigen B. Therefore, theoretically this type could be transfused into persons with blood of any other type. Individuals with type O blood are called universal donors. Type O blood, however, does contain both anti-A and anti-B antibodies. If type O blood is given to a person with blood type A, B, or AB, it should be transfused slowly so that the recipient’s larger blood volume will dilute the donor blood, minimizing the chance of an adverse reaction.

Figure 11. ABO Blood Types and Antibodies (different combinations of antigens and antibodies distinguish blood types)

ABO Blood Group

Table 1. Antigens and Antibodies of the ABO Blood Group

Blood Type

Antigen

Antibody

A

A

Anti-B

B

B

Anti-A

AB

A and B

Neither anti-A nor anti-B

O

Neither A nor B

Both anti-A and anti-B

Table 2. Preferred and Permissible Blood Types for Transfusions

Blood Type of Recipient

Preferred Blood Type of Donor

If Preferred Blood Type Unavailable, Permissible Blood Type of Donor

A

A

O

B

B

O

AB

AB

A, B, O

O

O

No alternate types

Rh Blood Group

The Rh (Rhesus) blood group was named after the rhesus monkey in which it was first studied. In humans, this group includes several Rh antigens (factors). The most prevalent of these is antigen D, a transmembrane protein.

If the Rh antigens are present on the red blood cell membranes, the blood is said to be Rh-positive. Conversely, if the red blood cells do not have Rh antigens, the blood is called Rh-negative. The presence (or absence) of Rh antigens is an inherited trait. Anti-Rh antibodies (anti-Rh) form only in Rh-negative individuals in response to the presence of red blood cells with Rh antigens. This happens, for example, if an individual with Rh-negative blood receives a transfusion of Rh-positive blood. The Rh antigens stimulate the recipient to begin producing anti-Rh antibodies. Generally, this initial transfusion has no serious consequences, but if an individual with Rh-negative blood—who is now sensitized to Rh-positive blood—receives another transfusion of Rh-positive blood some months later, the donated red cells are likely to agglutinate.

A similar situation of Rh incompatibility arises when an Rh-negative woman is pregnant with an Rh-positive fetus. Her first pregnancy with an Rh-positive fetus would probably be uneventful. However, if at the time of the infant’s birth (or if a miscarriage occurs) the placental membranes that separated the maternal blood from the fetal blood during the pregnancy tear, some of the infant’s Rh-positive blood cells may enter the maternal circulation. These Rh-positive cells may then stimulate the maternal tissues to produce anti-Rh antibodies. If a woman who has already developed anti-Rh antibodies becomes pregnant with a second Rh-positive fetus, these antibodies, called hemolysins, cross the placental membrane and destroy the fetal red blood cells. The fetus then develops a condition called erythroblastosis fetalis, or hemolytic disease of the fetus and newborn.

Erythroblastosis fetalis is extremely rare today because obstetricians carefully track Rh status. An Rh-negative woman who might carry an Rh-positive fetus is given an injection of a drug called RhoGAM at week 28 of her pregnancy and after delivery of an Rh-positive baby. Rhogam is a preparation of anti-Rh antibodies, which bind to and shield any Rh-positive fetal cells that might contact the woman’s cells and sensitize her immune system. RhoGAM must be given within 72 hours of possible contact with Rh-positive cells—including giving birth, terminating a pregnancy, miscarrying, or undergoing amniocentesis (a prenatal test in which a needle is inserted into the uterus).

References
  1. Anemia. National Heart, Lung and Blood Institute. https://www.nhlbi.nih.gov/health/health-topics/topics/anemia
  2. Iron-Deficiency Anemia. National Heart, Lung and Blood Institute. https://www.nhlbi.nih.gov/health-topics/topics/ida
  3. Pernicious Anemia. National Heart, Lung and Blood Institute. https://www.nhlbi.nih.gov/health/health-topics/topics/prnanmia
  4. Sickle Cell Disease. National Heart, Lung and Blood Institute. https://www.nhlbi.nih.gov/health/health-topics/topics/sca
  5. Who Is at Risk for Sickle Cell Disease ? National Heart, Lung and Blood Institute. https://www.nhlbi.nih.gov/health/health-topics/topics/sca/atrisk
  6. By en:User:Cburnett – Own work in Inkscape, CC BY-SA 3.0, https://commons.wikimedia.org/w/index.php?curid=1840082
  7. Polycythemia Vera. National Heart, Lung and Blood Institute. https://www.nhlbi.nih.gov/health/health-topics/topics/poly
read more
BloodImmune and Hematology System

What is blood plasma ?

blood plasma

What is blood plasma

How much blood you have depends mostly on your size and weight. A man who weighs about 70 kg (about 154 pounds) has about 5 to 6 liters of blood in his body. Blood is 55% blood plasma and about 45% different types of blood cells. Blood is composed of solid particles (red blood cells, white blood cells, and cell fragments called platelets) suspended in a fluid extracellular matrix called blood plasma. Over 99% of the solid particles present in blood are cells that are called red blood cells (erythrocytes) due to their red color. The rest are pale or colorless white blood cells (leukocytes) and platelets (thrombocytes).

The blood plasma is the clear, straw-colored, liquid portion of the blood in which the cells (red blood cells, white blood cells) and platelets are suspended. It is
approximately 92% water and less than 8% is dissolved substances, mostly proteins, a complex mixture of organic and inorganic biochemicals. Functions of plasma include transporting gases, vitamins, and other nutrients; helping to regulate fluid and electrolyte balance; and maintaining a favorable pH. Blood plasma also contain antibodies to fight infections (part of the immune system), glucose, amino acids and the proteins that form blood clots (part of the hemostatic system)..

Blood plasma contains electrolytes that are absorbed from the intestine or released as by-products of cellular metabolism. They include sodium, potassium, calcium, magnesium, chloride, bicarbonate, phosphate, and sulfate ions. Sodium and chloride ions are the most abundant. Bicarbonate ions are important in maintaining the pH of plasma. Like other plasma constituents, bicarbonate ions are regulated so that their blood concentrations remain relatively stable.

Blood transports a variety of materials between interior body cells and those that exchange substances with the external environment. In this way, blood helps maintain stable internal environmental conditions.

Hemostasis refers to the process that stops bleeding, which is vitally important when blood vessels are damaged. Following an injury to the blood vessels, several actions may help to limit or prevent blood loss. These include vascular spasm, platelet plug formation, and blood coagulation.

Platelets adhere to any rough surface, particularly to the collagen in connective tissue. When a blood vessel breaks, platelets adhere to the collagen underlying the endothelium lining blood vessels. Platelets also adhere to each other, forming a platelet plug in the vascular break. A plug may control blood loss from a small break, but a larger break may require a blood clot to halt bleeding.

Coagulation, the most effective hemostatic mechanism, forms a blood clot in a series of reactions, each one activating the next. Blood coagulation is complex and utilizes many biochemicals called clotting factors. Some of these factors promote coagulation, and others inhibit it. Whether or not blood coagulates depends on the balance between these two groups of factors. Normally, anticoagulants prevail, and the blood does not clot. However, as a result of injury (trauma), biochemicals that favor coagulation may increase in concentration, and the blood may coagulate. The resulting mass is a blood clot, which may block a vascular break and prevent further blood loss. The clear, yellow liquid that remains after the clot forms is called serum. Serum is plasma minus the clotting factors.

Note: Blood is a complex mixture of formed elements in a liquid extracellular matrix, called blood plasma. Note that water and proteins account for 99% of the blood plasma.

Figure 1. Blood composition

blood composition

blood compositionblood-composition

Note: Blood consists of a liquid portion called plasma and a solid portion (the formed elements) that includes red blood cells, white blood cells, and platelets. When blood components are separated by centrifugation, the white blood cells and platelets form a thin layer, called the “buffy coat,” between the plasma and the red blood cells, which accounts for about 1% of the total blood volume. Blood cells and platelets can be seen under a light microscope when a blood sample is smeared onto a glass slide.

Blood Plasma Proteins

Blood plasma proteins are the most abundant of the dissolved substances (solutes) in plasma. These proteins remain in the blood and interstitial fluids, and ordinarily are not used as energy sources. The three main types of blood plasma proteins—albumins, globulins, and fibrinogen—differ in composition and function.

Albumins are the smallest plasma proteins, yet account for about 60% of them by weight. Albumins are synthesized in the liver.

Plasma proteins are too large to pass through the capillary walls, so they are impermeant. They create an osmotic pressure that holds water in the capillaries, despite blood pressure forcing water out of capillaries by filtration. The term colloid osmotic pressure is used to describe this osmotic effect due to the plasma proteins. Because albumins are so plentiful, they are an important determinant of the colloid osmotic pressure of the plasma.

By maintaining the colloid osmotic pressure of plasma, albumins and other plasma proteins help regulate water movement between the blood and the tissues. In doing so, the blood plasma proteins help control blood volume, which, in turn, directly affects blood pressure.

If the concentration of plasma proteins falls, tissues swell. This condition is called edema. As the concentration of plasma proteins drops, so does the colloid osmotic pressure. Water leaves the blood vessels and accumulates in the interstitial spaces, causing swelling. A low plasma protein concentration may result from starvation, a protein-deficient diet, or an impaired liver that cannot synthesize plasma proteins.

Globulins make up about 36% of the plasma proteins. They can be further subdivided into alpha, beta, and gamma globulins. The liver synthesizes alpha and
beta globulins, which have a variety of functions. The globulins transport lipids and fat-soluble vitamins. Lymphatic tissues produce the gamma globulins, which are a type of antibody.

Fibrinogen constitutes about 4% of the plasma proteins, and functions in blood coagulation (clotting). Fibrinogen is synthesized in the liver, fibrinogen is the largest of the plasma proteins.

Plasma products and their uses

Plasma products can be grouped into three main types:

  • clotting or coagulation factors
  • albumin solutions
  • immunoglobulins

Coagulation or clotting factors

Coagulation is the name for the complex process of blood clotting. Clotting factors are proteins that work together with platelets to clot blood.

People need clotting factors for their blood to successfully clot. Missing one or more of these factors leads to blood clotting disorders such as haemophilia and Von Willebrand disease.

In the US, hemophilia is commonly treated with ‘recombinant factors’ that are manufactured in a laboratory and do not come from donated plasma.

Other blood clotting disorders that are treated with coagulation factors made from donated plasma.

Albumin

Albumin is the most common protein in blood plasma. It helps to:

  • carry substances around the body
  • maintain the right amount of fluid circulating in the body

If the circulation is working properly, vital hormones, cells and enzymes are transported to the right parts of the body to do their job.

If it’s not working properly, the circulatory system starts to break down, with serious consequences such as fluids being retained in the cells.

This can be treated by using human albumin solution which makes sure that the right amount of fluid is circulating in the blood stream.

Albumin can also be used to treat people with some types of liver or kidney disease and patients who have suffered burns.

Immunoglobulins

Immunoglobulins are protective antibodies which are produced by the body to fight against invading viruses or bacteria. There are two different types of immunoglobulins, specific and non-specific.

Specific immunoglobulins

Specific immunoglobulins contain high levels of antibody to a particular illness. These are given to people who have been exposed to a specific infection.

Antidotes to tetanus, rabies, chickenpox and hepatitis are all examples of specific immunoglobulins.

For example, a donor who has had chicken pox will have high levels of chicken pox antibodies. So their plasma would be ideal to treat a child with leukaemia who has been exposed to chicken pox.

Non-specific immunoglobulins

Non-specific immunoglobulins contain a wide variety of antibodies. These are given to people:

  • who make faulty antibodies, or can’t make their own antibodies
  • who are having treatments for diseases like cancer, where the treatment harms their ability to make antibodies

People with a faulty immune system need these products to live. Over 1,000 donations of plasma contribute to a single dose of an immunoglobulin product that contains all the necessary antibodies.

Table 1 summarizes the characteristics of the blood plasma proteins.

Table 1. Blood plasma proteins

Protein

Percentage of Total

Origin

Function

Albumins

60%

Liver

Help maintain colloid osmotic pressure

Globulins

36%

Alpha globulins

Liver

Transport lipids and fat-soluble vitamins

Beta globulins

Liver

Transport lipids and fat-soluble vitamins

Gamma globulins

Lymphatic tissues

Constitute the antibodies of immunity

Fibrinogen

4%

Liver

Plays a key role in blood coagulation

Separating plasma products

Plasma is first tested to be sure it’s safe to use, in the same way that whole blood is tested.

Many chemical and physical processes (e.g., spinning and heat treatments) are then carried out to separate the individual proteins. This is known as the fractionation process.

This process is fully automated and takes up to five days to complete.

Solvent or detergent treatment, dry heat treatment, filtration and pasteurization are also used to kill or remove any viruses that may be present.

The finished products are then tested again to make sure they contain the right biological make-up.

Once processing and testing is complete, the products are labeled, coded and packed, ready to be used by hospitals, clinics and doctors’ surgeries.

The total number of products that can be made with plasma fractionation runs into hundreds.

Blood Plasma Gases and Nutrients

The most important blood gases are oxygen (O2) and carbon dioxide (CO2). Blood plasma also contains a considerable amount of dissolved nitrogen, which ordinarily has no physiological function.

The plasma nutrients include amino acids, simple sugars, nucleotides, and lipids, all absorbed from the digestive tract. For example, blood plasma transports glucose from the small intestine to the liver, where it may be stored as glycogen or converted to fat. If blood glucose concentration drops below the normal range, glycogen may be broken down into glucose.

Blood plasma also carries recently absorbed amino acids to the liver, where they may be used to manufacture proteins, or deaminated and used as an energy source.

Blood plasma lipids include fats (triglycerides), phospholipids, and cholesterol. Because lipids are not water-soluble and plasma is almost 92% water, these lipids are carried in the plasma attached to proteins.

Blood Plasma Nonprotein Nitrogenous Substances

Molecules that contain nitrogen atoms but are not proteins comprise a group called nonprotein nitrogenous substances. In plasma, this group includes amino acids, urea, uric acid, creatine and creatinine. Amino acids come from protein digestion and amino acid absorption. Urea and uric acid are products of protein and nucleic acid catabolism, respectively. Creatinine results from the metabolism of creatine. In the skeletal muscle creatine is part of creatine phosphate in muscle tissue, where it stores energy in phosphate bonds.

Blood Plasma Electrolytes

Blood plasma contains electrolytes that are absorbed from the intestine or released as by-products of cellular metabolism. They include sodium, potassium, calcium, magnesium, chloride, bicarbonate, phosphate, and sulfate ions. Sodium and chloride ions are the most abundant. Bicarbonate ions are important in maintaining the pH of plasma. Like other plasma constituents, bicarbonate ions are regulated so that their blood concentrations remain relatively stable.

Blood plasma donation

Plasmapheresis is the standard procedure by which blood plasma is separated from whole blood and collected 1. Blood flows through a single needle placed in an arm vein, into a machine that contains a sterile, disposable plastic kit. The plasma is isolated and channeled out into a special bag, and red blood cells and other parts of the blood are returned to you through the same needle.

Is Plasmapheresis Safe ?

Absolutely. The machine and the procedure have been evaluated and approved by the Food and Drug Administration (FDA), and all plastics and needles coming into contact with you are used once and discarded 1. At no time during the procedure is the blood being returned to you detached from the needle in your arm, so there is no risk of returning the wrong blood to you.

Who Is Eligible to Participate in the AB Plasma Program ?

Donors must have blood group AB and must be male, because men lack plasma proteins (antibodies) directed against blood cell elements 1. Otherwise, eligibility for plasmapheresis procedures is the same as that for whole-blood donation. The interval between consecutive group AB plasmapheresis donations at National Institutes of Health Blood Bank is 1 month.

How Long Does Plasmapheresis Take ?

Plasmapheresis procedures take about 40 minutes, but you should allow another 20 minutes for staff to obtain your medical history 1.

What is blood plasma used for

Blood plasma is the pale yellow liquid part of whole blood. It is enriched in proteins that help fight infection (part of the immune system) and aid the blood in clotting (part of the hemostatic system). AB plasma is plasma collected from blood group AB donors. It is considered “universal donor” plasma because it is suitable for all recipients, regardless of blood group. Due to its value as a transfusion component, it is sometimes referred to as “liquid gold.”

Blood plasma products available in the United States include fresh frozen plasma and thawed plasma that may be stored at 33.8 to 42.8°F (1 to 6°C) for up to five days 2. Blood plasma contains all of the coagulation factors. Fresh frozen plasma infusion can be used for reversal of anticoagulant effects. Thawed plasma has lower levels of factors V and VIII and is not indicated in patients with consumption coagulopathy (diffuse intravascular coagulation) 3.

Blood plasma transfusion is recommended in patients with active bleeding and an International Normalized Ratio (INR) greater than 1.6, or before an invasive procedure or surgery if a patient has been anticoagulated 4, 5. Blood plasma is often inappropriately transfused for correction of a high INR when there is no bleeding. Supportive care can decrease high-normal to slightly elevated INRs (1.3 to 1.6) without transfusion of plasma. Table 2 gives indications for plasma transfusion 4, 5, 6.

Table 2. Indications for Transfusion of Blood Plasma Products

IndicationAssociated condition/additional information

International Normalized Ratio > 1.6

Inherited deficiency of single clotting factors with no virus-safe or recombinant factor available—anticoagulant factors II, V, X, or XI

Prevent active bleeding in patient on anticoagulant therapy before a procedure

Active bleeding

Emergency reversal of warfarin (Coumadin)

Major or intracranial hemorrhage

Prophylactic transfusion in a surgical procedure that cannot be delayed

Acute disseminated intravascular coagulopathy

With active bleeding and correction of underlying condition

Microvascular bleeding during massive transfusion

≥ 1 blood volume (replacing approximately 5,000 mL in an adult who weighs 155.56 lb [70 kg])

Replacement fluid for apheresis in thrombotic microangiopathies

Thrombotic thrombocytopenic purpura; hemolytic uremic syndrome

Hereditary angioedema

When C1 esterase inhibitor is unavailable

[Source 2]

Cryoprecipitate

Cryoprecipitate is prepared by thawing fresh frozen plasma and collecting the precipitate. Cryoprecipitate contains high concentrations of factor VIII and fibrinogen 2. Cryoprecipitate is used in cases of hypofibrinogenemia, which most often occurs in the setting of massive hemorrhage or consumptive coagulopathy. Indications for cryoprecipitate transfusion are listed in Table 3 7, 8. Each unit will raise the fibrinogen level by 5 to 10 mg per dL (0.15 to 0.29 μmol per L), with the goal of maintaining a fibrinogen level of at least 100 mg per dL (2.94 μmol per L) 8. The usual dose in adults is 10 units of pooled cryoprecipitate 3. Recommendations for dosing regimens in neonates vary, ranging from 2 mL of cryoprecipitate per kg to 1 unit of cryoprecipitate (15 to 20 mL) per 7 kg 7.

Table 3. Indications for Transfusion of Cryoprecipitate

Adults

Hemorrhage after cardiac surgery

Massive hemorrhage or transfusion

Surgical bleeding

Neonates

Anticoagulant factor VIII deficiency*

Anticoagulant factor XIII deficiency

Congenital dysfibrinogenemia

Congenital fibrinogen deficiency

von Willebrand disease*


*—Use when recombinant factors are not available.

[Source 2]

Blood plasma for patients undergoing surgery on the heart or blood vessels

Cardiovascular surgery includes many types of major surgery on the heart and major blood vessels, including procedures such as: heart valve replacements, coronary artery bypass grafts, aortic aneurysm repairs and corrections or congenital abnormalities of the heart. Cardiovascular surgery is associated with a significant risk of bleeding, with 8% of patients losing more than 2 ml/kg/hour of blood postoperatively 9. A number of features make patients undergoing cardiovascular surgery more likely to bleed 10, 11:

  • These patients may be taking drugs that predispose towards bleeding, such as aspirin or clopidogrel.
  • Patients undergoing major heart surgery will often require a cardiopulmonary bypass, where a circuit is formed by removing the heart from the circulation by passing a catheter into the aorta and the pulmonary artery while a cardiopulmonary bypass machine circulates blood round the body and ensures that it is adequately oxygenated. Heparin is used to prevent the cardiopulmonary bypass circuit from clotting. Heparin is an anticoagulant and can predispose patients to bleeding. When cardiopulmonary bypass is complete, heparin is neutralised with protamine.
  • Hypothermia and acidosis during the procedure may also contribute towards excess bleeding.
  • Dilution of clotting factors with administration of intravenous fluid; this is a particular problem in the paediatric setting.
  • When acute bleeding develops, clotting factors are consumed, resulting in a coagulopathy and predisposing the patient towards further bleeding.

In some cases these patients will have a clearly defined bleeding risk. They may already be haemorrhaging and, if this is the case, treatments to reduce bleeding would be considered therapeutic. Alternatively they may have abnormal blood results, such as a prolonged prothrombin time, suggesting that clotting factors may be deficient. Lastly, in some cases it may be presumed that a coagulopathy may develop and that prophylactic treatment before this event would reduce the risk of bleeding.

Treatment strategies to reduce bleeding include optimising surgical technique to minimise blood loss; antifibrinolytic agents such as tranexamic acid; careful monitoring and neutralisation of heparin; optimising the management of anticoagulant and antiplatelet drugs; and blood components such as fresh frozen plasma 12.

Fresh frozen plasma obtained from whole blood from blood donors, is a source of procoagulant factors, including fibrinogen and is used for either the treatment or prophylaxis of bleeding 13. Many audits indicate that patients undergoing major cardiac and vascular surgery receive a significant proportion of all clinical plasma transfusions. Some studies have reported wide variation in the use of clinical plasma for cardiac surgery and in critical care among centres within the same country 14.

Fresh frozen plasma contains a number of factors that help blood to clot. The risk of bleeding in open heart surgery or surgery on the main blood vessels in the body is high. Fresh frozen plasma is sometimes administered to these patients to reduce bleeding. It can be administered prophylactically (to prevent bleeding) or therapeutically (to treat bleeding). However, there are risks of side effects from fresh frozen plasma, such as severe allergic reactions or breathing problems 15.

However a 2015 Cochrane Review found no evidence for the efficacy of fresh frozen plasma for the prevention of bleeding in heart surgery and it found some evidence of an increased overall need for red cell transfusion in those treated with fresh frozen plasma 15. There were no reported adverse events due to fresh frozen plasma transfusion. Overall the evidence for the safety and efficacy of prophylactic fresh frozen plasma for cardiac surgery is insufficient. The trials focused on prevention of bleeding and did not address prevention of bleeding for patients with abnormal blood clotting or for the treatment of bleeding patients.

Blood plasma for critically ill patients

Plasma transfusions are a frequently used treatment for critically ill patients, and they are usually prescribed to correct abnormal coagulation tests and to prevent or stop bleeding 16. Plasma transfusions have been used since 1941 17. In 2008, 4,484,000 plasma units were transfused in patients in the United States 18. More than 10% of critically ill patients, both adults and children, receive a plasma transfusion during their hospital stay, making plasma transfusion a frequently used treatment modality 19, 20, 21. In current practice, plasma transfusions are widely used in critical care; they are administered most often to correct abnormal coagulation tests or to prevent bleeding 22.

In situations in which active bleeding is attributable to a coagulation factor deficiency, plasma transfusions can constitute a life-saving intervention by improving coagulation factor deficit 23, especially in patients requiring massive transfusion 24. Although plasma transfusions are frequently prescribed for critically ill patients, some of the reasons for their use are not supported by evidence from medical research. Some research has found an association of plasma transfusions with worse outcomes, and other studies have suggested that plasma transfusions do not help to return blood to its normal thickness 25.

Blood plasma for chronic inflammatory demyelinating polyradiculoneuropathy

Chronic inflammatory demyelinating polyradiculoneuropathy is a disease that causes progressive or relapsing and remitting weakness and numbness. At least one or two cases per 100,000 of the population and may be as high as 8.9 per 100,000 26. It is probably caused by an autoimmune process. Chronic inflammatory demyelinating polyradiculoneuropathy is an uncommon disease that causes weakness and numbness of the arms and legs. The condition may progress steadily or have periods of worsening and improvement. Although not proven, chronic inflammatory demyelinating polyradiculoneuropathy is generally considered to be an autoimmune disease caused by either humoral or cell-mediated immunity directed against myelin around individual nerve fibres or Schwann cell antigens which have not been identified 27. In severe cases, the disease affects the actual nerve fibres themselves. There has been debate as to whether people with the clinical features of an acquired demyelinating neuropathy and a systemic disease, such as cancer, diabetes mellitus, systemic lupus erythematosus, and other connective tissue diseases should be categorised as having chronic inflammatory demyelinating polyradiculoneuropathy 28.

Immunosuppressive or immunomodulatory drugs would be expected to be beneficial. According to Cochrane systematic reviews, three immune system treatments are known to help. These are corticosteroids (‘steroids’), plasma exchange (which removes and replaces blood plasma), and intravenous immunoglobulin (infusions into a vein of human antibodies). Moderate- to high-quality evidence from two small trials shows that plasma exchange provides significant short-term improvement in disability, clinical impairment, and motor nerve conduction velocity in chronic inflammatory demyelinating polyradiculoneuropathy but rapid deterioration may occur afterwards 29. Adverse events related to difficulty with venous access, use of citrate, and haemodynamic changes are not uncommon.

Blood plasma for generalised myasthenia gravis

Myasthenia gravis is an autoimmune disease caused by antibodies in the blood which attack the junctions (against the nicotinic acetylcholine receptor) between nerves and muscles they stimulate. Less than five per cent of patients have auto-antibodies to a muscle tyrosine kinase. Myasthenia gravis is characterised by weakness and fatigability of voluntary muscle, which changes over time. Acute exacerbations are life-threatening because they can cause swallowing difficulties or respiratory failure. Historically, with treatment – including thymectomy, steroids, and immunosuppressive drugs – after one to 21 (mean 12) years, 6% of patients went into remission, 36% improved, 42% were unchanged, and 2% were worse 30. In recent years, expert opinion has highlighted the greater efficacy of combined immunosuppressive treatments 31. A new subtype of myasthenia is associated with autoantibodies to a muscle specific kinase and these antibodies are also pathogenic on passive transfer 32.

Plasma exchange was introduced in 1976 as a short-term therapy for acute exacerbations of myasthenia gravis 33. It is thought to work because the exchange removes circulating anti-AChR (anti-acetylcholine receptor) antibodies. However, improvement has also been reported in so-called seronegative myasthenia gravis (where no anti-AChR antibodies can be detected) following plasma exchange 34. A symposium held in 1978 35, and numerous papers have recognised the short-term benefit of plasma exchanges 36. The use of repeated plasma exchange over a long period in refractory myasthenia gravis has also been reported 37. Plasma exchange is used worldwide for the treatment of myasthenia gravis but despite the published case series and the conferences of experts many questions remains unanswered concerning its efficacy for the treatment of chronic, more or less severe, myasthenia gravis as well as of myasthenic exacerbation or crisis and its efficacy in comparison with other treatments. Few randomised controlled trials have been published 38, 39. Plasma exchange removes these circulating auto-antibodies. Many case series suggest that plasma exchange helps to treat myasthenia gravis. However, two Cochrane Reviews 2002 40 and 2003 41, both found no adequate randomised controlled trials have been performed to determine whether plasma exchange improves the short- or long-term outcome for chronic myasthenia gravis or myasthenia gravis exacerbation. However, many case series studies convincingly report short-term benefit from plasma exchange in myasthenia gravis, especially in myasthenic exacerbation or crisis. In severe exacerbations of myasthenia gravis one randomised controlled trial did not show a significant difference between plasma exchange and intravenous immunoglobulin. Further research is need to compare plasma exchange with alternative short-term treatments for myasthenic crisis or before thymectomy and to determine the value of long-term plasma exchange for treating myasthenia gravis.

Blood plasma for the prevention of ovarian hyperstimulation syndrome

Ovarian hyperstimulation syndrome is an iatrogenic, serious and potentially fatal complication of ovarian stimulation which affects 1% to 14% of all in vitro fertilisation (IVF) or intracytoplasmic sperm injection (ICSI) cycles 42. Ovarian hyperstimulation syndrome may be associated with massive ovarian enlargement, extracellular exudate accumulation combined with profound intravascular volume depletion, ascites, hydrothorax, haemoconcentration, liver dysfunction and renal failure 43. It can lead to cancellation of an IVF cycle and prolonged bed rest or hospitalisation, which may have significant emotional, social, and economic impacts 44. Ovarian hyperstimulation syndrome can be classified into an early form that is related to the ovarian response and exogenous human chorionic gonadotrophin (hCG) administration, and is detected three to nine days after hCG administration. A late form of ovarian hyperstimulation syndrome, diagnosed 10 to 17 days later, is due to endogenous hCG 45 and is categorised as mild, moderate, severe or life-threatening. The aetiology of ovarian hyperstimulation syndrome is not completely clear at this moment; however the syndrome is strongly associated with serum hCG and certain vasoactive substances 46 are not elevated during gonadotropin stimulation in in vitro fertilization (IVF) patients developing ovarian hyperstimulation syndrome (OHSS): results of a prospective cohort study with matched controls. European Journal of Obstetrics, Gynecology, and Reproductive Biology 2001;96:196-201. https://www.ncbi.nlm.nih.gov/pubmed/11384807)).

Blood plasma albumin has both osmotic and transport functions. It contributes about 75% of the plasma oncotic pressure and administration of 50 g human albumin solution will draw more than 800 mL of extracellular fluid into the circulation within 15 minutes 47. It has been suggested that the binding and transport properties of human albumin play a major role in the prevention of severe ovarian hyperstimulation syndrome, as albumin may result in binding and inactivation of the vasoactive intermediates responsible for the pathogenesis of ovarian hyperstimulation syndrome. The osmotic function is responsible for maintaining the intra-vascular volume in the event of capillary leakage, thus preventing the sequelae of hypovolaemia, ascites and haemoconcentration 48. A 2016 Cochrane Review concluded that there is some eEvidence suggesting that the plasma expanders assessed in this review (human albumin, hydroxyethyl starch and mannitol) reduce rates of moderate and severe ovarian hyperstimulation syndrome in women at high risk. Adverse events appear to be uncommon, but were too poorly reported to reach any firm conclusions 48.

References
  1. AB Plasma Donor Program. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/donationtypes/ab_plasma.html
  2. Transfusion of Blood and Blood Products: Indications and Complications. Am Fam Physician. 2011 Mar 15;83(6):719-724. http://www.aafp.org/afp/2011/0315/p719.html
  3. King KE, Bandarenko N. Blood Transfusion Therapy: A Physician’s Handbook. 9th ed. Bethesda, Md.: American Association of Blood Banks; 2008:236.
  4. Practice parameter for the use of fresh-frozen plasma, cryoprecipitate, and platelets. Fresh-Frozen Plasma, Cryoprecipitate, and Platelets Administration Practice Guidelines Development Task Force of the College of American Pathologists. JAMA. 1994;271(10):777–781.
  5. Holland LL, Brooks JP. Toward rational fresh frozen plasma transfusion: the effect of plasma transfusion on coagulation test results. Am J Clin Pathol. 2006;126(1):133–139.
  6. Liumbruno G, Bennardello F, Lattanzio A, Piccoli P, Rossetti G; Italian Society of Transfusion Medicine and Immunohaematology (SIMTI) Work Group. Recommendations for the transfusion of plasma and platelets. Blood Transfus. 2009;7(2):132–150.
  7. Poterjoy BS, Josephson CD. Platelets, frozen plasma, and cryoprecipitate: what is the clinical evidence for their use in the neonatal intensive care unit? Semin Perinatol. 2009;33(1):66–74.
  8. Callum JL, Karkouti K, Lin Y. Cryoprecipitate: the current state of knowledge. Transfus Med Rev. 2009;23(3):177–188.
  9. Vuylsteke A, Pagel C, Gerrard C, Reddy B, Nashef S, Aldam P, et al. The Papworth Bleeding Risk Score: a stratification scheme for identifying cardiac surgery patients at risk of excessive early postoperative bleeding. European Journal of Cardiothoracic Surgery 2011;39(6):924-30. https://www.ncbi.nlm.nih.gov/pubmed/21094051
  10. Bevan DH. Cardiac bypass haemostasis: putting blood through the mill. British Journal of Haematology 1999;104(2):208-19. https://www.ncbi.nlm.nih.gov/pubmed/10050700
  11. Hartmann M, Sucker C, Boehm O, Koch A, Loer S, Zacharowski K. Effects of cardiac surgery on hemostasis. Transfusion Medicine Reviews 2006;20(3):230-41. https://www.ncbi.nlm.nih.gov/pubmed/16787830
  12. Davidson S. State of the art: how I manage coagulopathy in cardiac surgery patients. British Journal of Haematology 2014;164(6):779-89. https://www.ncbi.nlm.nih.gov/pubmed/24450971
  13. Desborough M, Stanworth S. Plasma transfusion for bedside, radiologically guided, and operating room invasive procedures. Transfusion 2012;52(Suppl 1):20S-9S. https://www.ncbi.nlm.nih.gov/pubmed/22578367
  14. Stanworth SJ, Grant-Casey J, Lowe D, Laffan M, New H, Murphy MF, et al. The use of fresh-frozen plasma in England: high levels of inappropriate use in adults and children. Transfusion 2011;51(1):62-70. https://www.ncbi.nlm.nih.gov/pubmed/20804532
  15. Desborough M, Sandu R, Brunskill SJ, Doree C, Trivella M, Montedori A, Abraha I, Stanworth S. Fresh frozen plasma for cardiovascular surgery. Cochrane Database of Systematic Reviews 2015, Issue 7. Art. No.: CD007614. DOI: 10.1002/14651858.CD007614.pub2. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD007614.pub2/full
  16. Plasma transfusion strategies for critically ill patients. National Center for Biotechnology Information, U.S. National Library of Medicine. https://www.ncbi.nlm.nih.gov/pubmedhealth/PMH0061586/
  17. Schmidt PJ. The plasma wars: a history. Transfusion 2012;52(S1):2S-4S.
  18. Office of the Assistant Secretary for Health. The 2009 National Blood Collection and Utilization Survey Report. Washington, DC: US Department of Health and Human Services, 2011.
  19. Luk C, Eckert KM, Barr RM, Chin-Yee IH. Prospective audit of the use of fresh-frozen plasma, based on Canadian Medical Association transfusion guidelines. Canadian Medical Association Journal 2002;166(12):1539-40. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC113799/
  20. Puetz J, Witmer C, Huang Y, Raffini L. Widespread use of fresh frozen plasma in US children’s hospitals despite limited evidence demonstrating a beneficial effect. Journal of Pediatrics 2012;160(2):210-5.
  21. Stanworth SJ, Walsh TS, Prescott RJ, Lee RJ, Watson DM, Wyncoll D. A national study of plasma use in critical care: clinical indications, dose and effect on prothrombin time. Critical Care 2011;15(2):R108.
  22. Vlaar AP, in der Maur AL, Binnekade JM, Schultz MJ, Juffermans NP. A survey of physicians’ reasons to transfuse plasma and platelets in the critically ill: a prospective single-centre cohort study. Transfusion Medicine 2009;19(4):207-12.
  23. Stanworth SJ, Hyde CJ, Murphy MF. Evidence for indications of fresh frozen plasma. Transfusion Clinique et Biologique 2007;14(6):551-6. https://www.ncbi.nlm.nih.gov/pubmed/18430602
  24. Zink KA, Sambasivan CN, Holcomb JB, Chisholm G, Schreiber MA. A high ratio of plasma and platelets to packed red blood cells in the first 6 hours of massive transfusion improves outcomes in a large multicenter study. American Journal of Surgery 2009;197(5):565-70. https://www.ncbi.nlm.nih.gov/pubmed/19393349
  25. Karam O, Tucci M, Combescure C, Lacroix J, Rimensberger PC. Plasma transfusion strategies for critically ill patients. Cochrane Database of Systematic Reviews 2013, Issue 12. Art. No.: CD010654. DOI: 10.1002/14651858.CD010654.pub2. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD010654.pub2/full
  26. Mahdi-Rogers M, Hughes RA. Epidemiology of chronic inflammatory neuropathies in southeast England. European Journal of Neurology 2014;21(1):28-33. https://www.ncbi.nlm.nih.gov/pubmed/23679015
  27. Köller H, Kieseier BC, Jander S, Hartung HP. Chronic inflammatory demyelinating polyneuropathy. New England Journal of Medicine 2005;352(13):1343-56. https://www.ncbi.nlm.nih.gov/pubmed/15800230
  28. Van den Bergh PY, Hadden RD, Bouche P, Cornblath DR, Hahn A, Illa I, et al. European Federation of Neurological Societies, Peripheral Nerve Society. European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of chronic inflammatory demyelinating polyradiculoneuropathy: report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society – first revision. Journal of the Peripheral Nervous System : JPNS 2010; Vol. 17, issue 3:356-63.
  29. Mehndiratta MM, Hughes RAC, Pritchard J. Plasma exchange for chronic inflammatory demyelinating polyradiculoneuropathy. Cochrane Database of Systematic Reviews 2015, Issue 8. Art. No.: CD003906. DOI: 10.1002/14651858.CD003906.pub4. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD003906.pub4/full
  30. Grob D, Brunner NG, Namba T. The natural course of myasthenia gravis and effect of therapeutic measures. Annals of the New York Academy of Science 1981;377(1):652-69. https://www.ncbi.nlm.nih.gov/pubmed/6951490
  31. Oosterhuis HJGH. Myasthenia gravis. Groningen: Groningen Neurological Press, 1997.
  32. Cole RN, Reddel SW, Gervasio OL, Phillips WD. Anti-MuSK patient antibodies disrupt the mouse neuromuscular junction.. Ann Neurol 2008;63(6):782-9. https://www.ncbi.nlm.nih.gov/pubmed/18384168
  33. Pinching AS, Peters DK. Remission of myasthenia gravis following plasma exchange. Lancet 1976;2(8000):1373-6. https://www.ncbi.nlm.nih.gov/pubmed/63848
  34. Miller RG, Milner-Brown HS, Dau PC. Antibody-negative acquired myasthenia gravis. Successful therapy with plasma exchange (letter). Muscle and Nerve 1981;4(3):255. https://www.ncbi.nlm.nih.gov/pubmed/7242562
  35. Dau PC. Plasmapheresis and the immunology of myasthenia gravis. Boston: Hougton-Mifflin, 1979.
  36. NIH Consensus Conference. The utility of therapeutic plasmapheresis for neurological disorders. NIH Consensus Development.. Journal of the American Medical Association 1986;256(10):1333-7. https://www.ncbi.nlm.nih.gov/pubmed/3747048
  37. Rodnitzky RL, Bosch EP. Chronic long-interval plasma exchange in myasthenia gravis. Archives of Neurology 1984;41(7):715-7. https://www.ncbi.nlm.nih.gov/pubmed/6743060
  38. Rønager J, Ravnborg M, Hermansen I, Vorstrup S. Immunoglobulin treatment versus plasma exchange in patients with chronic moderate to severe myasthenia gravis. Artificial Organs 2001;25(12):967-73. https://www.ncbi.nlm.nih.gov/pubmed/11843764
  39. Kamel A, Essa M. Effectiveness of prethymecthomy plasmapheresis on short-term outcome of non-thymomatous generalized myasthenia gravis. Egyptian Journal Neurology, Psychiatry and Neurosurgurgery 2009;46(1):161-8.
  40. Gajdos P, Chevret S, Toyka KV. Plasma exchange for generalised myasthenia gravis. Cochrane Database of Systematic Reviews 2002, Issue 4. Art. No.: CD002275. DOI: 10.1002/14651858.CD002275. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD002275/full
  41. Gajdos P, Chevret S, Toyka K. Intravenous immunoglobulin for myasthenia gravis. The Cochrane Database of Systematic Reviews 2003, Issue 2. Art. No.: CD002277. DOI: 10.1002/14651858.CD002277. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD002277
  42. Garcia-Velasco JA, Pellicer A. New concepts in the understanding of the ovarian hyperstimulation syndrome. Current Opinion in Obstetrics and Gynecology 2003;15(3):251-6. https://www.ncbi.nlm.nih.gov/pubmed/12858114
  43. Vloeberghs V, Peeraer K, Pexsters A, D’Hooghe T. Ovarianhyperstimulation syndrome and complications of ART. Best Practice & Research. Clinical Obstetrics & Gynaecology 2009;23(5):691-709. https://www.ncbi.nlm.nih.gov/pubmed/19632900
  44. Engmann L, DiLugie A, Schmidt D, Nulsen J, Maier D, Benavida C. The use of gonadotropin-releasing hormone (GnRH) agonist to induce oocyte maturation after cotreatment with GnRH antagonist in high-risk patients undergoing in vitro fertilization prevents the risk of ovarian hyperstimulation syndrome: a prospective randomised controlled study. Fertility and Sterility 2008;89(1):84-91. https://www.ncbi.nlm.nih.gov/pubmed/17462639
  45. Mathur RS, Jenkins JM. Is ovarian hyperstimulation syndrome associated with a poor obstetric outcome?. BJOG 2000;107:943-6. https://www.ncbi.nlm.nih.gov/pubmed/10955422
  46. Enskog A, Nilsson L, Brännström M. Plasma levels of free vascular endothelial growth factor(165) (VEGF(165
  47. McClelland DB. Human albumin solutions. BMJ 1990;300:35-57. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1661866/
  48. Shalev E, Giladi Y, Matilsky M, Ben-Ami M. Decreased incidence of severe ovarian hyperstimulation syndrome in high risk in-vitro fertilisation patients receiving intravenous albumin: a prospective study. Human Reproduction 1995;10:1373-6. https://www.ncbi.nlm.nih.gov/pubmed/7593499
read more
BloodImmune and Hematology System

Blood donation facts

blood donation

Blood donation rules

Blood donation rules and requirements vary a lot between countries. For example, in the the United States (US) you must be at least 17 years old to donate blood at the National Institutes of Health Blood Bank 1. In Australia the minimum age to donate blood is 16 years with an upper limit of 70 years of age 2. In the US there is no upper age limit for donation 1.

The temperature of blood in the body is 38° C, which is about one degree higher than body temperature 3. How much blood you have depends mostly on your size and weight. A man who weighs about 70 kg (about 154 pounds) has about 5 to 6 liters of blood in his body.

The volume of blood removed during a donation (450 mL ± 10% of blood) represents only about 10% of the total blood mass of a subject weighing 70 Kg.

Blood is 55% blood plasma and about 45% different types of blood cells. The blood plasma is a light yellow liquid. Over 90% of blood plasma is water, while less than 10% is dissolved substances, mostly proteins. Blood plasma also contains electrolytes, vitamins and nutrients such as glucose and amino acids. Over 99% of the solid particles present in blood are cells that are called red blood cells (erythrocytes) due to their red color. The rest are pale or colorless white blood cells (leukocytes) and platelets (thrombocytes).

Note: Blood is a complex mixture of formed elements in a liquid extracellular matrix, called blood plasma. Note that water and proteins account for 99% of the blood plasma.

Figure 1. Blood composition

blood composition

blood compositionblood-composition

Note: Blood consists of a liquid portion called plasma and a solid portion (the formed elements) that includes red blood cells, white blood cells, and platelets. When blood components are separated by centrifugation, the white blood cells and platelets form a thin layer, called the “buffy coat,” between the plasma and the red blood cells, which accounts for about 1% of the total blood volume. Blood cells and platelets can be seen under a light microscope when a blood sample is smeared onto a glass slide.

Blood has three important functions:

  • Transportation

The blood transports oxygen from the lungs to the cells of the body, where it is needed for metabolism. The carbon dioxide produced during metabolism is carried back to the lungs by the blood, where it is then exhaled. Blood also provides the cells with nutrients, transports hormones and removes waste products, which the liver, the kidneys or the intestine, for example, then get rid of.

  • Regulation

The blood helps to keep certain values of the body in balance. For instance, it makes sure that the right body temperature is maintained. This is done both through blood plasma, which can absorb or give off heat, as well as through the speed at which the blood is flowing. When the blood vessels expand, the blood flows more slowly and this causes heat to be lost. When the environmental temperature is low the blood vessels can contract, so that as little heat as possible is lost. Even the so-called pH value of the blood is kept at a level ideal for the body. The pH value tells us how acidic or alkaline a liquid is. A constant pH value is very important for bodily functions.

  • Protection

If a blood vessel is damaged, certain parts of the blood clot together very quickly and make sure that a scrape, for instance, stops bleeding. This is how the body is protected against losing blood. White blood cells and other messenger substances also play an important role in the immune system.

Blood donation eligibility 4

You can donate blood if you:

  • Are healthy
  • Are at least 17 years of age
  • Weigh at least 110 pounds (50 kgs). There is no upper weight limit.

The tables below address specific situations affecting donation eligibility.

Table 1. Medical Condition

Medical ConditionEligibility
Acquired Immune Deficiency Syndrome (AIDS), individuals at high risk and their partnersbanned 1cannot donate
Colds and flu within 2 daysbanned 1cannot donate
Diabetes, on or off medication and under control. If well-controlled by diet, oral medication, or insulin, you can donate.

*However, the use of insulin made from beef is a cause for permanent deferral.

tick1can donate
Hepatitis or jaundice after age 11banned 1cannot donate
Pregnancy. You cannot donate until six weeks after the conclusion of the pregnancy.banned 1cannot donate
Pregnancy, after delivery, miscarriage, abortiontick16-week wait
Menstruationtick1can donate
Cancer, treatment complete and disease-free; most typestick12-year wait
[Source 1]

Table 2. Medical Procedures

Medical ProceduresEligibility
Surgery, without transfusion (except for autologous blood)tick1can donate
Surgery, with transfusiontick11-year wait
[Source 1]

Table 3. Vaccinations

VaccinationsEligibility
Measles (rubella); measles, mumps, and rubella (MMR); chicken pox (varicella)tick11-month wait
Flutick1can donate
Hepatitis A or Btick1can donate
[Source 1]

Table 4. Other Possible Restrictions

Other Possible RestrictionsEligibility
Ear/Body piercing, sterile procedure/equipment usedtick1can donate
Ear/Body piercing, nonsterile procedure/equipment usedtick11-year wait
Tattooingtick11-year wait
Travel outside the United States or CanadaContact the National Institutes of Health Blood Bank
[Source 1]

If you have traveled to other countries

There is a slight risk of exposure to infectious agents outside the United States that could cause serious disease. Donor deferral criteria for travel outside the US are designed to prevent the transmission of three specific organisms from donor to recipient:

  • Malaria. Malaria is caused by a parasite that can be transmitted from mosquitoes to humans. It is found in several hundred countries, and is one of the leading causes of death from infectious diseases world-wide. Donors who have traveled to areas listed by the Centers for Disease Control 5 as malarial risk areas are deferred for 1 year after their travel ends 1.
  • Bovine Spongiform Encephalopathy (BSE). BSE is commonly referred to as “Mad Cow disease” and is caused by an abnormal, transmissible protein called a prion. In the 1990s, the United Kingdom experienced an epidemic of the disorder in cows, with subsequent cow-to-human transmission, presumably through the food chain. BSE-infected cattle were also detected in other countries in Western Europe. Transfusion-transmission of BSE among donor-recipient pairs has been documented in a handful of cases 1. Donors who have spent more than three months in the United Kingdom from 1980-1996, and donors who resided in Western Europe for greater than five years since 1980, are permanently deferred from blood donation 6.

Other blood donation restrictions

  • If you have low Hemoglobin (Hb) or anemia, female donors must have a hemoglobin level of at least 12.5g/dL and male donors are required to have a minimum level of 13.0g/dL. The deferral is 30 days for both whole blood and apheresis donations. The most common reason for low hemoglobin is iron deficiency, and you will be given information about maintaining a healthy iron balance.
  • If you have sickle cell disease, you cannot donate if you have sickle cell disease. You should not donate whole blood if you have sickle cell trait, because your blood will clog the filter that is applied to whole blood units. You can donate platelets if you are a sickle cell trait carrier.
  • If you have received a blood transfusion, you must wait one year to donate.
  • You cannot donate if you are currently experiencing severe allergy symptoms.
  • Taking antibiotics,  you can donate 24 hours after the last dose if you have no further signs of infection. You may donate while taking antibiotics for acne.
  • You cannot donate while taking narcotics to relieve pain.
  • You may donate blood while taking nonnarcotic pain relievers.
  • Aspirin interferes with platelet function and should be discontinued prior to platelet donation as follows. You cannot donate platelets if you have taken aspirin in the last 48 hours.
    • Special Caution: Many medications contain aspirin, so check the container carefully before making a platelet donation,
  • Nonaspirin: You can donate platelets if you have taken ibuprofen or other nonaspirin, nonsteroidal anti-inflammatory drugs (NSAIDS).
  • You can donate if you had skin cancer (basal cell or squamous cell) or cervical cancer in situ and the surgical site is completely healed.
  • If you had another type of cancer, you can donate two years after the date of surgery or other definitive therapy, as long as your doctor informs you that there is no evidence of persistent or recurrent cancer.
  • You are permanently deferred if you had leukemia or lymphoma.
  • If you have a cold or the flu, you can donate once you have been symptom-free for 48 hours.
  • If you have had dental work, there is a 24-hour waiting period after a routine cleaning and a 72-hour wait after extractions, root canals, or oral surgery.
  • If you have epilepsy, you can donate if you have been seizure free for at least one week.
  • If you have high blood pressure, you can donate if your blood pressure is controlled by medication.
  • If you have low blood pressure, you can donate with low blood pressure.
  • If you have heart disease or had a heart attack, in some situations, you may donate if you have heart disease or have had a heart attack. Contact the National Institutes of Health Blood Bank 7 for more details.
  • If you have had angioplasty, you will need to contact the National Institutes of Health Blood Bank 7 for more details.
  • If you had major surgery, you must wait until you have completely recovered and returned to normal activity before donating.
  • If you have had hepatitis, you cannot donate if you have had hepatitis after age 11.
  • If you received the hepatitis vaccine, you can donate if you have received the hepatitis vaccine (a series of three vaccinations).
  • You must wait one year if you received Hepatitis B Immune Globulin or if you experienced a needlestick injury contaminated with untested blood.
  • If you have herpes, you cannot donate with herpes or a cold sore when the lesions are active. You can donate if the lesions are dry and almost healed.
  • If you have had malaria, you may donate if you have been asymptomatic of malaria for more than 3 years while residing in a non-endemic country.

Benefits of blood donation

There is no substitute for human blood. Human blood cannot be manufactured. People are the only source of blood. Much of the medical care of an National Institutes of Health patient depends on the steady supply of blood from healthy, caring individuals like yourself. No miracle of modern medicine can help the patients who need blood if blood is not available. The blood you donate at the National Institutes of Health Blood Bank is used to support the many patients who come from all over the world to receive treatment.

Blood in the umbilical cord at birth is rich in stem cells that can be used to treat a variety of disorders, including leukemias, sickle cell disease and other hemoglobin abnormalities, and certain inborn errors of metabolism. The United States and the United Kingdom have public umbilical cord blood banks that provide stem cells for free. For many illnesses it is more effective to receive donor stem cells, because receiving one’s own could reintroduce the disease.

Types of Blood Donations

You can make the following types of donations:

Blood components for transfusion

  • Whole blood
  • Red cells by apheresis
  • Platelets by apheresis
  • Granulocytes by apheresis
  • AB Plasma by apheresis

Hemochromatosis donations

Hemochromatosis is a relatively common inherited condition in which the body absorbs excess iron. Over many years, iron overload can develop, with deposition of excess iron in body tissues and organs. Disabling arthritis, glandular failure, and severe liver disease can occur if the disorder is not treated. The treatment is phlebotomy therapy, or removal of 1 unit (1 pint) of blood every 1 – 16 weeks, depending on the level of iron overload. One pint of blood contains 250 mg of iron. Serial frequent phlebotomy sessions are a highly effective way to lower body iron levels. The National Institutes of Health Blood Bank has a protocol for treatment of hemochromatosis by phlebotomy therapy, which uses a simple and easy method to determine the pace of therapy.

The blood units removed therapeutically may be made available for transfusion into others if the donor (the hemochromatosis subject) meets standard blood donor eligibility criteria. Phlebotomy therapy and medical care for hemochromatosis are offered free of charge to all study participants. All persons with hemochromatosis are eligible for participation in this study, regardless of whether they meet blood donor criteria.

Hemochromatosis patients participating in the NIH Blood Bank Hemochromatosis Donor Program may donate:

  • Whole blood
  • Double red cells

Blood components for laboratory research use

Donated blood is never wasted. It is used every day of the year to treat patients who are participating in the medical treatment and research programs of the National Institutes of Health. If your blood is not required for immediate use, it may be frozen and stored. Fractions of blood that cannot be used for transfusion are used for research. Occasionally, the blood you give may not be required for a patient here but may be sent to another hospital, where it can be used to help save a life.

What is Whole Blood ?

Whole blood consists of red blood cells, white blood cells, and platelets suspended in a protective yellow liquid known as plasma. Most patients receiving transfusions do not need all of these elements.

Red blood cells have no nucleus, in contrast to many other cells. Each red blood cell contains hemoglobin, which can transport oxygen. In tiny blood vessels in the lung the red blood cells pick up oxygen from inhaled air and carry it through the bloodstream to all parts of the body. When they reach their goal, they release it again. The cells need oxygen for metabolism, which also creates carbon dioxide as a waste product. The red blood cells then pick up the carbon dioxide and transport it back to the lung. There we exhale it when we breathe out.

Red blood cells can also pick up or release hydrogen and nitrogen. When picking up or releasing hydrogen they help to keep the pH level of the blood steady; by releasing nitrogen the blood vessels expand, and blood pressure falls. Red blood cells have a life cycle of about 120 days. When they are too old or damaged they are broken down in the bone marrow, spleen or liver.

All solid parts of the blood originate from common parent cells, the so-called stem cells. In adults blood cells are produced mainly in the bone marrow. The various blood cells develop in several stages from stem cells to blood cells or blood platelets. White blood cells such as lymphocytes do not mature only in the bone marrow, but also in the lymph nodes. When the cells are completed, they are released into the bloodstream. In addition to these mature cells, the blood still contains a small number of precursor cells.

Figure 2. Blood cell development. A blood stem cell goes through several steps to become a red blood cell, platelet, or white blood cell

blood cell development

Certain messenger substances regulate the production of blood cells. The hormone erythropoietin, which is produced in the kidneys, promotes the production of red blood cells, while so-called cytokines stimulate the production of white blood cells.

One pint (473 ml) of whole blood is usually processed by a spinning method into:

  • Red blood cells, which carry the oxygen needed by patients who are anemic;
  • Platelets: Platelets are small cells that help the blood to clot. Manufactured in the bone marrow and stored in the spleen, their job is to rush to the site of an injury. Once there, they form a barrier, help the damaged organ or blood vessel stop bleeding, and give the body a chance to begin healing. Platelets needed by patients who are bleeding.
  • Plasma: Plasma is the pale yellow liquid part of whole blood, in which the cellular elements are suspended. It is enriched in proteins that help fight infection and aid the blood in clotting. AB plasma is plasma collected from blood group AB donors. It is considered “universal donor” plasma because it is suitable for all recipients, regardless of blood group. Due to its value as a transfusion component, it is sometimes referred to as “liquid gold.” Plasma is transfused to patients whose blood is not clotting.

Therefore, one donation of whole blood can treat three different patients!

Used for:

  • Cancer, blood diseases, haemophilia, anaemia, heart disease, stomach disease, kidney disease, childbirth, operations, blood loss, trauma, burns.

Is it Safe to Give Whole Blood ?

Absolutely. Many safeguards are taken to assure that no harm comes to you during or after donation. First, we obtain a medical history and check your blood pressure, pulse, and temperature to assure that you are in good health. We take a small sample of blood from your finger to confirm that you have an ample number of red cells to share. All equipment used to collect your blood is sterile and disposable. After donation, we provide delicious snacks and drinks.

Who is Eligible to Give Whole Blood?

Please see detailed description of donor criteria above (Tables 1-4 plus eligibility criteria and restrictions).

How often can you donate whole blood ?

You must wait 56 days between whole-blood donations to allow the number of red blood cells in your body to return to a predonation level.

How long does blood donation take ?

15 minutes to donate, 45 minutes for the appointment.

How Do You Arrange to Donate ?

You can contact the National Institutes of Health Blood Bank 7 for more details.

What is Donating Red Cells by Apheresis

Double Red Cell Apheresis (DRCA) is a procedure that allows a donor to give 2 units of red cells at the same time. This is done by a procedure called “apheresis,” which separates whole blood into component parts such as red blood cells, platelets, and plasma 8. To remove red cells, a needle is placed in your arm, and the blood flows into a sterile, disposable plastic kit installed in a machine designed specifically for this purpose. As blood enters the machine, the bowl is spinning at high speed. This causes the components of the blood to separate so that the red blood cells can be siphoned into a blood bag. Plasma and other parts of the blood are then returned to you through the same needle. The process is repeated to collect 2 units of red blood cells.

Is Double Red Cell Apheresis Safe ?

Absolutely. The machine and the procedure have been evaluated and approved by the Food and Drug Administration (FDA), and all plastics and needles coming into contact with you are used once and discarded 8. At no time during the procedure is the blood being returned to you detached from the needle in your arm, so there is no risk of returning the wrong blood to you.

What Are The Double Red Cell Apheresis Donor Requirements ?

Because you will be losing more red blood cells than usual during this procedure, donor criteria differ from those for whole blood donation:

Men

  • Minimum weight – 130 pounds (59 kgs)
  • Minimum height – 5’1″

Women

  • Minimum weight – 150 pounds (68 kgs)
  • Minimum height – 5’5″

Donors must have a slightly higher red cell count, specifically, a fingerstick hemoglobin level of at least 13.3 gm/dL. To assure that you do not become anemic, the interval between Double Red Cell Apheresis and subsequent blood donation is 4 months 8.

How Long Does Double Red Cell Apheresis Procedure Take ?

The time required to remove 2 units of reds cells is about 45 minutes 8. Because you’ll be with us longer, there will be time to watch TV, read, or just chat with staff and other donors. Every effort will be made to make the experience relaxing and enjoyable.

What is Donating Platelets by Apheresis

Plateletpheresis is the standard procedure by which platelets are separated from whole blood, concentrated, and collected 9. To remove platelets, a needle is placed in each arm. Blood flows through a needle into a machine that contains a sterile, disposable plastic kit specifically designed for this purpose. The platelets are isolated and channeled out into a special bag, and red blood cells and other parts of the blood are returned to you through a needle in the opposite arm. There is also a plateletpheresis procedure that can be performed with a single needle.

Is Plateletpheresis Safe ?

Absolutely. The machine and the procedure have been evaluated and approved by the Food and Drug Administration, and all plastics and needles coming into contact with you are used once and discarded 9. At no time during the procedure is the blood being returned to you detached from the needle in your arm, so there is no risk of returning the wrong blood to you.

Who Is Eligible to Give Platelets ?

The interval between consecutive platelet donations at National Institutes of Health Blood Bank is 1 month. However, because the body replaces platelets within a few days, you are allowed to give more frequently when you are donating for a relative or for a patient who responds particularly well to your platelets. In addition to standard donor eligibility requirements, platelet donors should refrain from taking aspirin for 48 hours prior to donation.

How Long Does Plateletpheresis Take ?

Plateletpheresis procedures take about 90 minutes 9, but you should allow another 30 minutes for staff to obtain your medical history.

What is Plasmapheresis ?

Plasmapheresis is the standard procedure by which plasma is separated from whole blood and collected 10. Blood flows through a single needle placed in an arm vein, into a machine that contains a sterile, disposable plastic kit. The plasma is isolated and channeled out into a special bag, and red blood cells and other parts of the blood are returned to you through the same needle.

Is Plasmapheresis Safe ?

Absolutely. The machine and the procedure have been evaluated and approved by the Food and Drug Administration (FDA), and all plastics and needles coming into contact with you are used once and discarded 10. At no time during the procedure is the blood being returned to you detached from the needle in your arm, so there is no risk of returning the wrong blood to you.

Who Is Eligible to Participate in the AB Plasma Program ?

Donors must have blood group AB and must be male, because men lack plasma proteins (antibodies) directed against blood cell elements 10. Otherwise, eligibility for plasmapheresis procedures is the same as that for whole-blood donation. The interval between consecutive group AB plasmapheresis donations at National Institutes of Health Blood Bank is 1 month.

How Long Does Plasmapheresis Take ?

Plasmapheresis procedures take about 40 minutes, but you should allow another 20 minutes for staff to obtain your medical history 10.

What is Granulocytes by Apheresis

A granulocyte is a type of white blood cell which fights bacteria and fungi. Patients with bone marrow failure syndromes, or those undergoing chemotherapy or marrow transplantation, often do not make enough of their own granulocytes to prevent serious infections. Such patients may benefit from a course of granulocyte transfusions.

What is Granulocytapheresis ?

Granulocytapheresis is the process by which granulocytes are collected from a healthy donor 11. Similar to plateletpheresis, blood is withdrawn from a vein and directed into a machine that contains a sterile disposable kit. Granulocytes are separated by a spinning process and concentrated in a plastic bag. The remaining parts of the blood are returned to the donor through a second needle.

Is Granulocytapheresis Safe ?

Yes. The machine and the procedure used for granulocyte collection are approved by the Food and Drug Administration, and all plastics and needles coming into contact with the donor are discarded after use 11. Two medications are given to the donor the day before collection to boost the donor’s granulocyte count. These medications are approved for donor use under an NIH protocol.

Who Is Eligible to Give Granulocytes ?

Granulocyte donors are healthy, unpaid volunteers between the age of 18 and 75 who meet the criteria for blood and platelet donors described below 11. Granulocyte donors must meet some additional criteria described in the protocol. Granulocyte donors should refrain from taking aspirin for 48 hours prior to donation.

How Long Does Granulocytapheresis Take ?

Giving granulocytes with use of an apheresis machine takes about 2.5 hours, which is slightly longer than a plateletpheresis donation 11. The donor has enough time to enjoy a full-length movie.

INCLUSION CRITERIA for Granulocytapheresis:

  • Donors shall meet all donor eligibility criteria for allogeneic blood donors, as defined in the most recent editions of the AABB Standards and FDA Code of Federal Regulations (21CFR640).

In addition, donors shall meet the following restrictions:

  • Age greater than or equal to 18 and less than or equal to 75 years
  • If hypertension is present, must be well-controlled on medications
  • If peptic ulcer disease has been diagnosed in the past, symptoms must be well-controlled on medications
  • If cataracts have been diagnosed in the past, records from subject’s ophthalmologist must be obtained indicating type of cataract. If posterior subcapsular cataract was diagnosed in the past, subject may receive G-CSF but not dexamethasone. The only exception to this is a history of bilateral cataract extractions due to posterior subcapsular cataract.

EXCLUSION CRITERIA for Granulocytapheresis:

  • Information obtained from health history screen that does not meet the allogeneic donor eligibility criteria of the AABB Standards or the FDA CFR.
  • Weight less than 50 kg (110 lbs)
  • History of coronary heart disease
  • Uncontrolled hypertension (systolic BP >160, diastolic BP >100)
  • History of hepatitis or injection drug use
  • Diabetes mellitus requiring insulin
  • Active, symptomatic peptic ulcer disease
  • History of iritis or episcleritis
  • Sickle cell disease (sickle trait is acceptable). Testing for hemoglobin S is not required.
  • Lithium therapy
  • Pregnancy or nursing (breast feeding)

Blood donation side effects

Blood donors normally tolerate the donation very well, but occasionally adverse reactions of variable severity may occur during or at the end of the collection. The adverse reactions that occur in donors can be divided into local reactions and systemic reactions. In a small study involving 4,906 donors (3,716 male and 1,190 female), only 63 donors (1.2% of all the volunteers) suffered some kind of adverse reaction: 59 (1.08% of the subjects) had mild reactions (agitation, sweating, pallor, cold feeling, sense of weakness, nausea), and only 4 (3 males and 1 female, 0.2%) had more severe disorders, including vomiting, loss of consciousness, and convulsive syncope.

The statistical probability of an episode of complete loss of consciousness was equivalent to an incidence of 0.1%, that is, one case every 1,000 donors 12.

Of the 63/4,906 (1.2%) donors who had adverse reactions to blood donation, only 40 (0.8%) were administered oral vasopressors (a drug or other agent which causes the constriction of blood vessels), while the other 23 (0.4%) recovered from the vasovagal reaction simply from being placed in the antishock position. Of the 63 donors who had a vasovagal reaction, only 18 (0.36%) were given crystalloid solutions, while less than five (0.1%) required additional therapy with cortisone 12. In no case was re-animation based on oxygen therapy or administration of adrenaline needed.

The greatest number of reactions occurred during or after donations of whole blood, with there being fewer after donations of other blood components.

Local reactions

Local reactions occur predominantly because of problems related to venous access 12. They are usually haematomas due to extravasation from the veins, caused by incorrect placement of the needle during the venipuncture. Pain, hyperaemia and swelling may develop at the site of the extravasation. Other local events include pain due to slight trauma to the subcutaneous nerve endings. In most cases, however, these are banal complications that do not require any treatment. Local phlebitis and thrombophlebitis are more serious complications than the foregoing, but are very rare.

Systemic reactions

The systemic reactions, in contrast to the local reactions, can be divided into mild or severe. In most cases, they are vasovagal reactions than can be triggered by the pain of the venipuncture, by the donor seeing his or her own blood, by the donor seeing another donor unwell, by the anxiety and state of tension of undergoing the donation, etc. The systemic reactions are characterised by the appearance of pallor, sweating, dizziness, gastrointestinal disorders, nausea, hypotension, and bradycardia. Therapeutic intervention must be swift, otherwise this clinical picture, typical of a vasovagal reaction, will progress into an episode of syncope, of variable severity, which may or may not be complicated by the onset of tonic-clonic muscle spasms (convulsive syncope), accompanied by vomiting and loss of sphincter control.

Systemic reactions can occur during apheresis procedures, which require the use of anticoagulants such as acid-citrate-dextrose for the collection of the blood component 12. This anticoagulant can cause hypocalcaemia, because of chelation. The lowered concentration of calcium ions leads to episodes of paraesthesia of the lips, oral cavity and limbs. These symptoms resolve after interruption of the apheresis procedure, although it may sometimes be necessary to use a therapeutic intervention, such as the administration of calcium gluconate. Much more rarely, tremor, muscle spasms, hypotension, tachycardia, arrhythmia, convulsions and tetany develop.

There are rare reports of acute intoxication due to overdoses of acid-citrate-dextrose 13. Another rare complication, that can occur during apheresis procedures, is severe arrhythmias 12.

Figure 3. Frequency of the symptoms occurring in donors during or immediately after the donation

symptoms during or after blood donation

Note: The donor population analysed consisted of 4,906 donors (3,716 male and 1,190 female). In total, 3,983 (81%) voluntaries have donated whole blood, 851 (17%) plasma from apheresis, 64 (1.3%) experienced multicomponent donation, and 8 (0.1%) were donors of plasma-platelet apheresis in the year from 24th October, 2005 to 24th April 2006.

[Source 12]

Blood Transfusions

Early attempts to transfer blood from one person to another produced varied results. Sometimes, the recipient improved. Other times, the recipient suffered a blood transfusion reaction in which the red blood cells clumped, obstructing vessels and producing great pain and organ damage.

Eventually scientists determined that blood is of differing types and that only certain combinations of blood types are compatible. These discoveries led to the development of procedures for typing blood. Today, safe transfusions of whole blood depend on two blood tests—“type and cross match.” First the recipient’s ABO blood type and Rh status are determined. Following this, a “cross match” is done of the recipient’s serum with a small amount of the donor’s red blood cells that have the same ABO type and Rh status as the recipient. Compatibility is determined by examining the mixture under a microscope for agglutination, the clumping of red blood cells. Agglutination (“clumping”) is a reaction between antigens and specific antibodies. This is what happens when antigens on mismatched donated red blood cells react with antibodies in plasma.

Blood Antigens and Antibodies

An antigen, is any molecule that triggers an immune response, the body’s reaction to invasion by a foreign substance or organism. When the immune system encounters an antigen not found on the body’s own cells, it will attack, producing antibodies. In a transfusion reaction, antigens (agglutinogens) on the surface of the donated red blood cells react with antibodies (agglutinins) in the plasma of the recipient, resulting in the agglutination of the donated red blood cells.

A mismatched blood transfusion quickly produces telltale signs of agglutination—anxiety, breathing difficulty, facial flushing, headache, and severe pain in the neck, chest, and lumbar area. Red blood cells burst, releasing free hemoglobin. Liver cells and macrophages phagocytize the hemoglobin, converting it to bilirubin, which may sufficiently accumulate to cause the yellow skin of jaundice. Free hemoglobin reaching the kidneys may ultimately cause them to fail.

Only a few of the 32 known antigens on red blood cell membranes can produce serious transfusion reactions. These include the antigens of the ABO group and those of the Rh group. Avoiding the mixture of certain kinds of antigens and antibodies prevents adverse transfusion reactions.

ABO Blood Group

The ABO blood group is based on the presence (or absence) of two major antigens on red blood cell membranes—antigen A and antigen B. A and B antigens are carbohydrates attached to glycolipids projecting from the red blood cell surface. A person’s erythrocytes have on their surfaces one of four antigen combinations: only A, only B, both A and B, or neither A nor B.

A person with only antigen A has type A blood. A person with only antigen B has type B blood. An individual with both antigens A and B has type AB blood. A person with neither antigen A nor B has type O blood. Thus, all people have one of four possible ABO blood types—A, B, AB, or O. The resulting ABO blood type is inherited. It is the consequence of DNA encoding enzymes to synthesize A or B antigens on erythrocytes in one of these four combinations.

  • In the United States, the most common ABO blood types are O (47%) and A (41%). Rarer are type B (9%) and type AB (3%). These percentages vary in subpopulations and over time, reflecting changes in the genetic structure of populations.

Antibodies that affect the ABO blood group antigens are present in the plasma about two to eight months following birth, as a result of exposure to foods or microorganisms containing the antigen(s) that are not present on the individual’s red blood cells. Specifically, whenever antigen A is absent in red blood cells, an antibody called anti-A is produced, and whenever antigen B is absent on cells, an antibody called anti-B is produced. Therefore, individuals with type A blood have anti-B antibody in their plasma; those with type B blood have anti-A antibody; those with type AB blood have neither antibody; and those with type O blood have both anti-A and anti-B antibodies (Figure 4 and Table 5). The antibodies anti-A and anti-B are large and do not cross the placenta. Thus, a pregnant woman and her fetus may be of different ABO blood types, but agglutination in the fetus will not occur.

An antibody of one type will react with an antigen of the same type and clump red blood cells; therefore, such combinations must be avoided. The major concern in blood transfusion procedures is that the cells in the donated blood not clump due to antibodies in the recipient’s plasma. For this reason, a person with type A (anti-B) blood must not receive blood of type B or AB, either of which would clump in the presence of anti-B in the recipient’s type A blood. Likewise, a person with type B (anti-A) blood must not receive type A or AB blood, and a person with type O (anti-A and anti-B) blood must not receive type A, B, or AB blood.

Type AB blood does not have anti-A or anti-B antibodies, so an AB person can receive a transfusion of blood of any other type. For this reason, individuals with type AB blood are called universal recipients. However, type A (anti-B) blood, type B (anti-A) blood, and type O (anti-A and anti-B) blood still contain antibodies (either anti-A and/or anti-B) that could agglutinate type AB cells if transfused rapidly.

Consequently, even for AB individuals, using donor blood of the same type as the recipient is best (Table 6). Type O blood has neither antigen A nor antigen B. Therefore, theoretically this type could be transfused into persons with blood of any other type. Individuals with type O blood are called universal donors. Type O blood, however, does contain both anti-A and anti-B antibodies. If type O blood is given to a person with blood type A, B, or AB, it should be transfused slowly so that the recipient’s larger blood volume will dilute the donor blood, minimizing the chance of an adverse reaction.

Figure 4. ABO Blood Types and Antibodies (different combinations of antigens and antibodies distinguish blood types)

ABO Blood Group

Table 5. Antigens and Antibodies of the ABO Blood Group

Blood Type

Antigen

Antibody

A

A

Anti-B

B

B

Anti-A

AB

A and B

Neither anti-A nor anti-B

O

Neither A nor B

Both anti-A and anti-B

Table 6. Preferred and Permissible Blood Types for Transfusions

Blood Type of Recipient

Preferred Blood Type of Donor

If Preferred Blood Type Unavailable, Permissible Blood Type of Donor

A

A

O

B

B

O

AB

AB

A, B, O

O

O

No alternate types

Rh Blood Group

The Rh (Rhesus) blood group was named after the rhesus monkey in which it was first studied. In humans, this group includes several Rh antigens (factors). The most prevalent of these is antigen D, a transmembrane protein.

If the Rh antigens are present on the red blood cell membranes, the blood is said to be Rh-positive. Conversely, if the red blood cells do not have Rh antigens, the blood is called Rh-negative. The presence (or absence) of Rh antigens is an inherited trait. Anti-Rh antibodies (anti-Rh) form only in Rh-negative individuals in response to the presence of red blood cells with Rh antigens. This happens, for example, if an individual with Rh-negative blood receives a transfusion of Rh-positive blood. The Rh antigens stimulate the recipient to begin producing anti-Rh antibodies. Generally, this initial transfusion has no serious consequences, but if an individual with Rh-negative blood—who is now sensitized to Rh-positive blood—receives another transfusion of Rh-positive blood some months later, the donated red cells are likely to agglutinate.

A similar situation of Rh incompatibility arises when an Rh-negative woman is pregnant with an Rh-positive fetus. Her first pregnancy with an Rh-positive fetus would probably be uneventful. However, if at the time of the infant’s birth (or if a miscarriage occurs) the placental membranes that separated the maternal blood from the fetal blood during the pregnancy tear, some of the infant’s Rh-positive blood cells may enter the maternal circulation. These Rh-positive cells may then stimulate the maternal tissues to produce anti-Rh antibodies. If a woman who has already developed anti-Rh antibodies becomes pregnant with a second Rh-positive fetus, these antibodies, called hemolysins, cross the placental membrane and destroy the fetal red blood cells. The fetus then develops a condition called erythroblastosis fetalis, or hemolytic disease of the fetus and newborn.

Erythroblastosis fetalis is extremely rare today because obstetricians carefully track Rh status. An Rh-negative woman who might carry an Rh-positive fetus is given an injection of a drug called RhoGAM at week 28 of her pregnancy and after delivery of an Rh-positive baby. Rhogam is a preparation of anti-Rh  antibodies, which bind to and shield any Rh-positive fetal cells that might contact the woman’s cells and sensitize her immune system. RhoGAM must be given within 72 hours of possible contact with Rh-positive cells—including giving birth, terminating a pregnancy, miscarrying, or undergoing amniocentesis (a prenatal test in which a needle is inserted into the uterus).

References
  1. NIH BLOOD BANK. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/can_i_donate.html
  2. Age – How does age affect my ability to donate ? Australian Red Cross Blood Service. http://donateblood.com.au/faq/age
  3. What does blood do ? National Center for Biotechnology Information, U.S. National Library of Medicine. https://www.ncbi.nlm.nih.gov/pubmedhealth/PMH0072576/
  4. About Donating Blood. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/about_donating_blood.html
  5. Explore Travel Health with the CDC Yellow Book. Centers for Disease Control and Prevention. https://wwwnc.cdc.gov/travel/page/yellowbook-home
  6. Bovine Spongiform Encephalopathy (BSE) Questions and Answers. U.S. Food and Drug Administration. https://www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/ucm111482.htm
  7. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/index.html
  8. Red Cells by Apheresis. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/donationtypes/red_cells.html
  9. Platelets by Apheresis. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/donationtypes/platelets.html
  10. AB Plasma Donor Program. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/donationtypes/ab_plasma.html
  11. Granulocytes by Apheresis. National Institutes of Health Blood Bank. https://clinicalcenter.nih.gov/blooddonor/donationtypes/granulocytes.html
  12. Crocco A, D’Elia D. Adverse reactions during voluntary donation of blood and/or blood components. A statistical-epidemiological study. Blood Transfusion. 2007;5(3):143-152. doi:10.2450/2007.0005-07. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2535889/
  13. Winters JL. Complications of donor apheresis. J Clin Apher. 2006;21:132–41. https://www.ncbi.nlm.nih.gov/pubmed/15880355
read more
Health Jade