Immunopathology


Return to the tutorial menu.

Immune reactions are divided into two broad categories:

Humoral immunity

  • B-cell lymphocyte mediated via production of antibody
  • Often develops as a response to soluble antigens
  • B cells account for about 20% of circulating lymphocytes
  • Immunoglobulin gene rearrangements allow tremendous diversity of responses to many antigens

In the diagram of an immunoglobulin molecule above, note that there are two heavy (H) chains and two light (L) chains linked by disulfide bonds. Each heavy and light chain has a constant (C) and a variable (V) region. It is the variable regions in the Fab portion that react with a specific antigen and give rise to the diversity of immunologic response. Immunoglobulin can attach via the Fc portion to a variety of cells with Fc receptors.

Cellular immunity

  • T-cell lymphocyte mediated
  • CD4+ helper lymphocytes: help B cells make antibody and also help to generate cytotoxic T cells; participate in delayed hypersensitivity reactions; account for 60% of peripheral T lymphocytes
  • CD8+ suppressor lymphocytes: are cytotoxic; represent about 30% of circulating T lymphocytes
  • T-cell receptors (TCR) are genetically programmed to recognize specific antigens and can rearrange their alpha and beta genes to respond to antigenic stimuli
    • Macrophages process antigen and present it with class II HLA to the CD4+ cells
    • Cytokines such as interleukin (IL) and tumor necrosis factor (TNF) are elaborated by activated T cells to enhance cellular immune reactions

HISTOCOMPATIBILITY ANTIGENS

Major histocompatibility complex (MHC) or HLA complex is on chromosome 6

Class I antigens: A, B, and C; these are present on all nucleated cells and are linked to beta-2 microglobulin; they are tested for and detected by serologic means; in practice the "C" antigens are unimportant; a number of alleles are present, and each person inherits one from each parent; thus, a person might be HLA typed as:

A 5, 10
B 11, 41

Class II antigens are in the D region (DR) and have a narrow distribution (mostly on mononuclear inflammatory cells); they are detected by mixed lymphocyte assay

Why are HLA important?

  • HLA matching is important in transplantation
  • HLA regulate some immune responses
  • Virus-infected cells with class I antigen are lysed by CD8+ cells that can recognize the virus-cell complex
  • Class II antigens help to induce CD4+ cells
  • HLA are associated with a variety of diseases, such as HLA B27 with ankylosing spondylitis, or HLA DR 2, DR3, and DR4 with autoimmune diseases

HYPERSENSITIVITY REACTIONS

Type I hypersensitivity

Anaphylaxis: Prior sensitization has resulted in an immune response initially mediated by CD4 lymphocytes (of the Th2 variety) that promote mast cell proliferation and plasma cell production of IgE. The IgE becomes bound to mast cells in places such as respiratory tract mucosa. Encountering the allergen again leads to mast cell degranulation with release of primary mediators (such as histamine, serotonin) which cause vasodilation, bronchoconstriction, etc. and release of secondary mediators (such as leukotrienes, prostaglandin) which lead to inflammatory cell infiltrates. The process of mast cell degranulation is diagrammed below:

There are two forms of anaphylaxis:

  • Systemic anaphylaxis: In some individuals, a severe reaction occurs within minutes, leading to symptomatology such as acute asthma, laryngeal edema, diarrhea, urticaria, and shock. Classic examples are penicillin allergy and bee sting allergy.
  • Local anaphylaxis (atopy): About 10% of people have "atopy" and are easily sensitized to allergens that cause a localized reaction when inhaled or ingested. This can produce hay fever, hives, asthma, etc. Classic examples are food allergies and hay fever to ragweed pollen.

Type II Hypersensitivity

Complement dependent reactions: Antibody is directed against antigen on cells (such as circulating red blood cells) or extracellular materials (basement membrane). The resulting Ag-Ab complexes activate complement (via the classic pathway), leading to cell lysis or extracellular tissue damage.

In the above diagram, a red blood cell has antigen fixed on its surface to which antibody attaches. The attached antibody sets off the complement cascade, which ends with the formation of the "membrane attack complex" of C5-9 which causes lysis of the cell. Other complement components may be generated, such as the opsonin C3b.

Diseases in this complement dependent category include:

  • Transfusion reactions: incompatible RBC's or serum is transfused.
  • Autoimmune hemolytic anemia: antibody is made against one's own RBC's.
  • Erythroblastosis fetalis: maternal IgG crosses the placenta and attaches to fetal RBC's.
  • Goodpasture's syndrome: glomerular basement membrane antibody is present.

Antibody-dependent cell-mediated cytotoxicity (ADCC): Low concentrations of IgG or IgE (in the case of parasites) coat target cells. Inflammatory cells such as NK (natural killer) cells, monocytes, and granulocytes then bind to the immunoglobulin Fc receptors and lyse, but do not phagocytize, the target cells.

In the diagram above, a macrophage with Fc receptors on its surface is able to recognize a target cell coated with antibody via the Fc receptor portion of the attached antibody. The macrophage can then demolish the targeted cell by elaboration of proteases.

Examples of ADCC include:

  • Transplant rejection
  • Immune reactions against neoplasms
  • Immune reactions against parasites

Antireceptor antibodies: IgG antibody is directed against receptors in target cells, resulting in complement-mediated destruction of the receptors.

In the diagram above, antibody is directed against acetylcholine receptors at the motor end plate of a muscle, blocking the receptors and diminishing the muscular response. This is the mechanism for muscle weakness in myasthenia gravis.

Diseases caused by this mechanism include:

  • Myasthenia gravis: acetylcholine receptor antibody.
  • Grave's disease (thyrotoxicosis): anti-TSH receptor antibody
  • Pernicious anemia: anti-parietal cell antibody.

Type III Hypersensitivity

This reaction is mediated by immune (Ag-Ab) complexes which promote tissue damage primarily through complement activation (alternate pathway). C3b as an opsonin attracts neutrophils, which then release lysosomal enzymes. C5a as a chemoattractant brings in neutrophils. Serum complement is reduced as it is used up in this process.

In the diagram above, antigen-antibody complexes are circulating and becoming trapped beneath the basement membrane of a small blood vessel, setting off the complement cascade and generating components that attract PMN's to generate an ongoing inflammatory response.

Immune complexes can be deposited systemically or locally

Systemic immune complex disease: Ag-Ab complexes form in the circulatory system and are deposited in tissues, typically near basement membranes in places such as blood vessels, glomeruli, skin, joints, pleura, and pericardium. Larger immune complexes are quickly phagocytized by macrophages and removed, but small to intermediate complexes formed with antigen excess may escape removal leading to:

  • Glomerulonephritis
  • Serum sickness
  • Vasculitis

Local immune complex disease: Also called an "Arthus" reaction, it occurs with local injection of the antigen and leads to focal vasculitis. This kind of immune reaction also plays a role in the development of hypersensitivity pneumonitis (so-called "farmer's lung").

Type IV Hypersensitivity

This reaction is called "delayed hypersensitivity" because it is mediated by sensitized CD4+ T lymphocytes which process antigens in association with class II HLA molecules and release lymphokines. The lymphokines promote a reaction (especially mediated through macrophages) beginning in hours but reaching a peak in 2 to 3 days.

This diagram above illustrates a sequence of events in granuloma formation in response to Mycobacterium tuberculosis (MTB). The key cell in the process is the epithelioid macrophage.

Hypersensitivity reactions with this mode of action include:

  • Granulomatous diseases (mycobacteria, fungi)
  • Tuberculin skin reactions
  • Transplant rejection
  • Contact dermatitis

Cytotoxic T lymphocyte (CTL) mediated responses: CD8+ T cells are generated and lyse specific cells. Class I HLA molecules play a role. Reactions with this mode include:

  • Neoplastic cell lysis
  • Transplant rejection
  • Virus-infected cell lysis

TRANSPLANT REJECTION

Immunologic Mechanisms

  • The HLA system is a key factor in most reactions. Reactions are mediated by either T lymphocytes or by antibody. The major types of hypersensitivity reactions involved are types II and IV.
  • The ABO system, best characterized as the major blood group antigens, is also important because these antigens are expresed on all cells except those in the central nervous system. Thus, matching for ABO compatibility is important for transplantation.
  • T-cell mediated reactions: Can be either CD4+ cells generating delayed hypersensitivity reactions after recognizing foreign HLA class II (DR) antigens or cytotoxic CD8+ cells recognizing foreign HLA class I (A,B, or C) antigens. The donor tissue or donor lymphocytes within the transplanted tissue carry the offending HLA antigens.
  • Antibody mediated reactions: These can be mediated through complement-mediated cytotoxicity, antibody-dependent cytotoxicity (ADCC), or immune complexes.

Organ System Pathology

  • Renal transplants: three classic modes of rejection are described. HLA matching for both class I and class II antigens improves survival:
    • Hyperacute rejection: previous sensitization through transfusion, pregnancy, or infections (through HLA cross-reacting bacterial or viral antigens) leads to preformed antibody that causes immediate (minutes to hours) vascular injury via ADCC.
    • Acute rejection: Cellular infiltrates with both CD4+ and CD8+ cells are present. Occurence is sometimes within days, but usually within months (and sometimes years later when immunosuppressive therapy is discontinued). Humoral immunity is also involved in acute rejection, and necrotizing vasculitis occurs; intimal proliferation results. Ag-Ab complexes play a role.
    • Chronic rejection: There is intimal fibrosis with vascular thickening, leading to ischemic changes. Mononuclear infiltrates with prominent plasma cells are present. Both T-cell and humoral mechanisms are involved.

  • Liver transplants: HLA is less important than simple matching of organ size (since most of these are done in children). There are two modes of rejection:
    • Acute rejection: seen within two months, there are mixed inflammatory portal and central vein infiltrates.
    • chronic rejection: at some later time, with continued inflammation, portal fibrosis, arteriolar thickening, and bile ductular necrosis occurs.

  • Heart transplants: HLA is less important than simple matching of organ size. Immunosuppressive therapy is carefully monitored in relationship to signs of rejection seen on endomyocardial biopsy. There are two modes of rejection:
    • Acute cellular rejection: there are lymphocytic infiltrates and possible myocardial fiber necrosis.
    • Acute vascular rejection: immunoglobulin deposition occurs in small arteries and produces a vasculitis.

  • Bone marrow transplants: HLA matching is important. A distinct problem is graft versus host disease (GVHD) which results from the donor lymphocytes attacking the recipient tissues having the offending HLA antigens. Chemotherapy agents used to prepare the patient for marrow transplantation may result in hepatic veno-occlusive disease in the weeks following transplantation.

AUTOIMMUNE DISEASES

"The fancier the plumbing, the easier it is to stop up the drain" -- The human immune system is very complex and, hence, there are numerous ways it can malfunction. There are a number of diseases that can result, and many of these have similarities. It is sometimes difficult to separate them. They are also often called "connective tissue" disorders because many of them are manifested in a variety of tissues.

Hypersensitivity reactions involved in autoimmunity are primarily types II and III, though type IV reactions can also be present.

Mechanisms proposed for development of autoimmunity include:

Bypass of CD4+ T-cell tolerance of "self" antigens by:

  • complex of antigen with a hapten (such as a drug or infectious agent) or infectious degradation of an antigen
  • cross-reaction with a hapten on an infectious agent that is similar to tissue proteins--classic for poststreptoccocal glomerulonephritis and rheumatic heart disease
  • direct activation of B-cells via bacterial endotoxin and via Epstein-Barr virus receptors on B-cells

Idiotype bypass through ligand mimicry, as seen in antireceptor antibody mediated disease, and cross-reactivity with infectious agents:

  • T-suppressor/helper imbalance
  • emergence of a sequestered antigen through tissue trauma or inflammatory destruction. Examples include: lens crystalline of eye, spermatozoa in testis, and myelin in CNS

Antinuclear Antibodies

Many autoimmune diseases have serologic evidence for antibodies directed at components of "self" cell nuclei. Though these antibodies may not be the direct cause of, or evidence for, tissue injury, they are very useful for diagnosis and categorization of autoimmune diseases. The types include:

ANA (antinuclear antibody): seen in many autoimmune diseases and not diagnostic of any. In general, the higher the titer, the worse the disease. There are some characteristic fluorescent staining patterns for ANA:

  • homogenous (diffuse) - not very specific for anything
  • rim - may be indicative of anti-double stranded DNA, seen in SLE
  • speckled - indicative of antibody to extractable nuclear antigens, often seen in MCTD
  • nucleolar - antibody to nucleolar RNA, seen often in PSS
  • centromere - antibody to centromeric protein, seen in CREST syndrome

Though no autoantibody is completely sensitive or specific for a particular autoimmune disease, some of the strongest associations include:

  • Anti-double stranded DNA (native DNA antibody): SLE
  • Anti-Smith: SLE
  • anti-histone: drug-induced SLE
  • anti SS-A and anti SS-B: Sjogren's syndrome
  • anti DNA-topoisomerase I (Scl-70): PSS
  • anti-histidyl-tRNA synthetase (Jo-1): polymyositis
  • anti-RNP (ribonucleoprotein): MCTD
  • anti-phospholipid antibody (anti-cardiolipin antibody): SLE and others

Systemic Lupus Erythematosus (SLE)

The manifestations are protean, and there is no characteristic or pathognomonic finding. Instead, this disease is diagnosed by finding suggestive serologic and clinical findings. Findings may include:

  • Skin rash - malar or discoid
  • Sensitivity to light (photodermatitis)
  • Serositis - inflammation of serosal surfaces along with effusions
  • Glomerulonephritis - the worst problem with SLE
  • Cytopenias - anemia, leukopenia, thrombocytopenia
  • Antinuclear antibody - rim pattern, anti double stranded-DNA and anti-Smith autoantibodies are most specific for SLE
  • Arthralgias, myalgias
  • Vasculitis - anywhere: CNS, skin, kidney, etc
  • Decreased serum complement - especially C1q
  • Thrombosis - in arteries or veins

Genetic factors: tends to run in families; association with HLA Dr-2 and Dr-3; more common in young women (especially African-American).

Drugs can produce "drug-induced" SLE: list includes procainamide, hydralazine, isoniazid, d-penicillamine.

Discoid lupus erythematosus (DLE): a benign disease with skin involvement; ANA positive in only a third (but some of these go on to SLE).

Mixed Connective Tissue Disease (MCTD)

A wastebasket category for patients who do not clearly fit into other categories. There are features similar to SLE, scleroderma, and polymyositis. Most patients are middle aged females. Characteristic feature is a speckled ANA pattern.

Scleroderma (Progressive Systemic Sclerosis, PSS)

Characterized by excessive fibrosis in a variety of tissues from collagen deposition by activated fibroblasts. About 75% of cases are in women, mostly middle aged. Patterns of disease include:

Limited scleroderm, or CREST syndrome: the benign form of PSS, serologically suggested by the presence of anti-centromere antibody

  • C = Calcinosis in skin and elsewhere
  • R = Raynaud's phenomenon, sensitivity to cold
  • E = Esophageal dysmotility from submucosal fibrosis
  • S = Sclerodactyly from dermal fibrosis
  • T = Telangiectasias

Diffuse scleroderma: the worst form of PSS; Scl-70 (anti-DNA topoisomerase I) antibody shows specificity for this form. It may include all of the findings with CREST, but additionally has renal findings: arterial intimal thickening and proliferation (hyperplastic arteriolosclerosis) leading to malignant hypertension with arterial fibrinoid necrosis, thrombosis, and renal infarction. Half of diffuse PSS patients die from renal disease. The lungs in this form of scleroderma may have diffuse alveolar fibrosis leading to honeycomb fibrosis

Morphea: this is skin fibrosis only

Polymyositis-Dermatomyositis

Characterized by inflammation of skeletal muscle with weakness. Sometimes it is associated with a skin rash (hence, dermatomyositis). It is seen in ages 40-60, but also in ages 5-15, mostly in women. Some of these patients have Jo-1 antibody. Inflammation in polymyositis is mainly mediated by cytotoxic CD8 cells. In dermatomyositis, mainly antigen-antibody complexes produce a vasculitis in muscle and skin. Some adults (10-20%) develop cancer.

Sjogren's Syndrome

Characterized by dry eyes and dry mouth as a result of lacrimal and salivary gland involvement by lymphocytic infiltration, fibrosis, and destruction mediated by CD4+ cells helping antibody production, of which anti-SS-A and anti-SS-B are the most specific. Most patients are middle to older age women. Lacrimal and salivary gland inflammation of any cause (including Sjogren's) is called Mikulicz's syndrome.

PRIMARY IMMUNODEFICIENCY SYNDROMES

X-linked Agammaglobulinemia of Bruton

Congenital agammaglobulinemia (Bruton's Disease) results from a genetic defect on the long arm of X chromosome, so that males are primarily affected (inheritance occurs in an X-lined recessive pattern). The mutations affect production of a tyrosine kinase (Bruton tyrosine kinase, or btk) active in early pre-B cells which diminishes their maturation and leads to virtual absence of all immunoglobulin classes. Infants are observed to have multiple infections with bacterial organisms (Hemophilus, Staphylococcus), particularly in skin and lung. Agammaglobulinemia is the result of absent B-cells, but T-cell mediated immunity is intact. If affected persons survive, many will develop autoimmune diseases.

Common Variable Immunodeficiency

This is a heterogenous group of disorders with an incidence of 1 per 100,000 that can involve both humoral and cell mediated immunity. Though there are normal numbers of circulating B lymphocytes, there is impaired secretion of one or more immunoglobulin isotypes, usually IgG or IgA. A selective abnormality of T cell activation, as demonstrated by decreased synthesis of interleukins (IL 2, 4, and 5) has been identified. Patients may have impaired gastrointestinal mucosal immunity. Another variant results from either a decrease in CD4 cells or an increase in CD8 cells. Also occuring is a variant resulting from the presence of T and B lymphocyte autoantibodies.

At least two of the three main serum immunoglobulin isotypes are decreased. Persons with CVID are prone to recurrent bacterial infections, particularly sinusitis, bronchitis, pneumonia, bronchiectasis, and otitis. Bordatella pertussis infections occur in childhood. Viral infections are uncommon, though recurrent herpes simplex with eventual herpes zoster is an exception. Giardiasis is common. Half of CVID cases are diagnosed before age 21, but in some cases complications do not develop until adolescence or adulthood. There is an increased incidence of autoimmune diseases, particularly hemolytic anemia, thrombocytopenia, and pernicious anemia. In about two thirds of cases, normal numbers of circulating B lymphocytes are present. There is a decrease in immunoglobulins, generally in all classes, more often IgG and IgA, but sometimes only of IgG.

DiGeorge Syndrome

The DiGeorge syndrome (or sequence) is a field defect of third and fourth pharyngeal pouch development in utero during organogenesis in the first trimester of pregnancy. A specific deletion on the long arm of chromosome 22 has been implicated. Anatomic structures that may be aplastic or hypoplastic include the thymus, parathyroid glands, great vessels, and esophagus. DiGeorge syndrome may be further subclassified as complete, in which there is almost total absence of thymic tissue, or as partial, in which there is only a decrease in thymic tissue

Complete DiGeorge syndrome is characterized by normal levels of circulating immunoglobulin, though in some cases serum IgE is increased and IgA is decreased. However, affected children have markedly decreased numbers of circulating T lymphocytes, making them susceptible to fungal and viral infections. Children with partial DiGeorge syndrome have only a slight decrease in peripheral T lymphocytes and have increased infections, but with less frequency and with less severity than children with the complete form. Accompanying aplasia of parathyroid glands can lead to life-threatening hypocalcemia that may appear soon after birth.

Severe Combined Immunodeficiency (SCID)

SCID results from different defects with different inheritance patterns, but all demonstrate some degree of failure in development of both humoral and cell-mediated immunity. The major variants of SCID include:

  • An X-linked form is due to a mutation on the long arm of the X chromosome which produces a defective gamma chain of the interleukin-2 receptor (cytokine receptor). Lacking an intact interleukin receptor renders early lymphocytes incapable of normal differentiation and development to functional T and B cells in response to growth factors. This form accounts for about 60% of cases.
  • Autosomal recessive inheritance linked to a lack of the enzyme adenosine deaminase (ADA) leads to about 35 to 40% of cases. The ADA enzyme is involved in purine metabolism, and its deficiency results in production of metabolites toxic to lymphocytes

There tends to be a greater decrease in cell mediated immunity than in humoral immunity. Normal or increased numbers of B lymphocytes may be present with the X-linked form, but these cells still do not function properly. There is very little serum IgG and virtually no IgM or IgA. Infants develop Candida skin rashes and thrush, persistent diarrhea, severe respiratory tract infections with Pneumocystis carinii (jirovecii) and Pseudomonas soon after birth, and failure to thrive after 3 months of age. Severe viral can occur. Maternal T lymphocytes crossing the placenta may produce graft versus host disease

Wiskott-Aldrich Syndrome

An X-linked recessive pattern is seen because the defective gene is located on the short arm of the X chromosome (Xp11.23). The immunodeficiency is accompanied by thrombocytopenia and eczema. Circulating platelets are markedly decreased. T lymphocytes exhibit cytoskeletal disorganization and loss of microvilli by electron microscopy, and they express little CD43 by immunohistochemical staining.

There is usually a normal level of serum IgG, along with a decrease in IgM, but often an increase in both IgA and IgE. The initial onset of disease in early childhood is accompanied by recurrent bacterial infections, particularly to encapsulated bacteria such as Streptococcus pneumoniae, with development of pneumonia, meningitis, and septicemia. Later, failure of T lymphocyte function may predispose to recurrent herpetic infections and to Pneumocystis carinii (jirovecii) pneumonia. A bleeding problem may result from the severe thrombocytopenia.

Ataxia-Telangiectasia

A genetic defect is present on the long arm of chromosome 11 which predisposes to chromosome breakage and rearrangement, particularly on chromosomes 7 and 14, leading to a high risk for neoplasia and a marked sensitivity to radiation. This disorder is quite rare and has an autosomal recessive pattern of inheritance.

There is a triad of progressive cerebellar ataxia, mucocutaneous telangiectasias, and recurrent respiratory tract infections with a variety of bacterial and fungal organisms. Immunoglobulin deficiencies, particularly IgA and/or IgE, may be present, though serum IgM is usually elevated. The symptoms usually begin between 9 months and 2 years of age.

Selective IgA Deficiency

About 1 in 600 persons of European descent has a virtual lack of circulating IgA as well as secretory IgA which, in most cases, results from failure of the IgA type of B lymphocytes to transform into plasma cells capable of producing IgA or from impaired survival of IgA producing plasma cells. Some patients may have deficiencies in IgG subclasses 2 and/or 4, while IgG subclasses 1 and 3 are increased. Some patients go on to develop common variable immunodeficiency, suggesting that there is a similar defect in B cell maturation and function.

Some affected patients are at increased risk for respiratory, gastrointestinal, and urinary tract infections, most often bacterial, and diarrhea is common. More severely affected persons may even have a sprue-like illness with malabsorbtion. Atopy, as demonstrated by asthma, can be present. Concomitant autoimmune diseases, particularly systemic lupus erythematosus and rheumatoid arthritis, can be present. About half of IgA deficient persons develop anti-IgA antibodies of the IgE type, so that transfusion of blood products containing serum with normal IgA levels leads to severe systemic anaphylaxis.

Other Primary Immunodeficiency Disorders

Deficiencies of other components of the immune system are uncommon. Some of the best known are:

  • Complement component deficiencies: C2 deficiency carries a risk for development of autoimmune disease. C3 deficiency is associated with recurrent bacterial infections.
  • Chediak-Higashi syndrome: A rare autosomal recessive disorder in which peripheral blood neutrophils, monocytes, and lymphocytes contain giant cytoplasmic granules and patients have leukopenia, making them susceptible to bacterial and fungal infections of skin, mucous membranes, and respiratory tract
  • Chronic granulomatous disease: neutrophils and monocytes lack the enzyme NADPH oxidase which is needed to generate intracellular oxidants that destroy phagocytosed infectious organisms, particularly catalase-positive agents such as Staphylococcus aureus, Candida, and Aspergillus, so that chronic infections are common

Return to the tutorial menu.