Vi Disease of The Immune System-7150
Vi Disease of The Immune System-7150
The main mechanisms of innate immunity and adaptive immunity.NK cells, Natural killer cells
(from Robbins and Cotran; Pathologic basis of disease)
Innate immunity is always present, ready to provide defense against microbes and to eliminate damaged cells. The
receptors and components of innate immunity have evolved to serve these purposes. Innate immunity functions in stages:
recognition of microbes and damaged cells, activation of various mechanisms, and elimination of the unwanted
substances.The major components of innate immunity are epithelial barriers that block entry of microbes, phagocytic cells
(mainly neutrophils and macrophages), dendritic cells,natural killer (NK) cells, and several plasma proteins, including the
proteins of the complement system.Epithelia of the skin and gastrointestinal and respiratory tracts provide mechanical
barriers to the entry of microbes from the external environment. Epithelial cells also produce antimicrobial molecules such
as defensins, and lymphocytes located in the epithelia combat microbes at these sites. If microbes do breach epithelial
boundaries, other defense mechanisms are called in. In addition to immune cells, several soluble proteins play important
roles in innate immunity. The proteins of the complement system are plasma proteins that are activated by microbes using
the alternative and lectin pathways in innate immune responses; in adaptive immunity it is activated by antibodies using the
classical pathway. Other circulating proteins of innate immunity are mannose-binding lectin and C-reactive protein, both of
which coat microbes and promote phagocytosis (opsonins). Lung surfactant is also a component of innate immunity,
providing protection against inhaled microbes.
The adaptive immune system consists of lymphocytes and their products, including antibodies. The receptors of
lymphocytes are much more diverse than those of the innate immune system, but lymphocytes are not inherently specific for
microbes, and they are capable of recognizing a vast array of foreign substances. In the remainder of this introductory section
we focus on lymphocytes and the reactions of the adaptive immune system.There are two types of adaptive immunity:
humoral immunity, which protects against extracellular microbes and their toxins, and cell-mediated (or cellular) immunity,
which is responsible for defense against intracellular microbes. Humoral immunity is mediated by B (bone marrow–derived)
lymphocytes and their secreted products, antibodies (also called immunoglobulins, Ig), and cellular immunity is mediated
by T (thymus-derived) lymphocytes. Both classes of lymphocytes express highly specific receptors for a wide variety of
substances, called antigens.
IMMUNE CELLS. The principal cells of the immune system are the lymphocytes, antigen-presenting cells, and
effector cells. Lymphocytes are the cells that specifically recognize and respond to foreign antigens. Accessory cells, such
as macrophages and dendritic cells, function as antigen-presenting cells by the processing of a complex antigen into epitopes
required for the activation of lymphocytes. Functionally, there are two types of immune cells: regulatory cells and effector
cells. The regulatory cells assist in orchestrating and controlling the immune response. For example, helper T lymphocytes
activate other lymphocytes and phagocytes. The final stages of the immune response are accomplished with the elimination
of the antigen by effector cells. Activated T lymphocytes, mononuclear phagocytes, and other leukocytes function as effector
cells in different immune responses.
Lymphocytes represent 25% to 35% of blood leukocytes, and 99% of the lymphocytes reside in the lymph. Like
other blood cells, lymphocytes are generated from stem cells in the bone marrow. Undifferentiated immature lymphocytes
congregate in the central lymphoid tissues, where they develop into distinct types of mature lymphocytes. One class of
lymphocyte, the B lymphocytes (B cells), matures in the bone marrow and is essential for humoral, or antibody-mediated,
immunity. The other class of lymphocyte, the T lymphocytes (T cells), completes its maturation in the thymus and functions
in the peripheral tissues to produce cell-mediated immunity, as well as aiding antibody production. Approximately 60% to
70% of blood lymphocytes are T cells, and 10% to 20% are B cells. The various types of lymphocytes are distinguished by
their function and response to antigen, their cell membrane molecules and receptors, their types of secreted proteins, and
their tissue location. High concentrations of mature T and B lymphocytes are found in the lymph nodes, spleen, skin, and
mucosal tissues, where they can respond to antigen.
The key trigger for the activation of B and T cells is the recognition of the antigen by unique surface receptors.
The B-cell antigen receptor consists of membrane-bound immunoglobulin molecules that can bind a specific epitope.
The T-cell receptor recognizes a processed antigen peptide in association with a self-recognition protein, called a
major histocompatibility complex (MHC) molecule. The appropriate recognition of MHC and self peptides or MHC
associated with for n peptides is essential for lymphocytes to differentiate “self” from “foreign.”
The immune system enlists specialized antigen-presenting cells (APCs), such as macrophages and dendritic cells,
to ensure the appropriate processing and presentation of antigen. On recognition of antigen and after additional stimulation
by various secreted signaling molecules called cytokines, the B and T lymphocytes divide several times to form populations
or clones of cells that continue to differentiate into several types of effector and memory cells. These effector cells and
molecules defend the body in an immune response. In humoral or antibody-mediated immunity, activated B cells produce
effector cells called plasma cells, which secrete protein molecules called antibodies, or immunoglobulins. The binding of
antibodies can neutralize the biologic impact of the microbes and cause subsequent aggregation to ensure their removal.
Phagocytic cells can more efficiently bind, engulf, and digest antigen–antibody aggregates or immune complexes than they
can antigen alone.
T and B cells display additional membrane molecules called clusters of differentiation (CD) molecules. These
molecules aid the function of immune cells and also serve to define functionally distinct subsets of cells, such as CD4+
helper T cells and CD8+ cytotoxic T cells. The many cell surface CD molecules detected on immune cells have allowed
scientists to identify distinct subsets of lymphocytes and study both the normal and abnormal developmental processes
displayed by these cells. In cell-mediated immunity, regulatory CD4+ helper T cells enhance the response of other T cells,
and effector cytotoxic T lymphocytes (CD8+) destroy cellular antigens such as tumor cells and virus-infected cells.
T and B lymphocytes possess all of the key properties associated with the adaptive immune response—specificity,
diversity, memory, and self- and nonself-recognition. These cells can exactly recognize a particular microorganism or foreign
molecule. Each lymphocyte targets a specific antigen and differentiates that invader from other substances that may be
similar. The approximately 1012 lymphocytes in the body have tremendous diversity. They can respond to the millions of
different kinds of antigens encountered daily. This diversity occurs because an enormous variety of lymphocyte populations
have been programmed during development, each to respond to a different antigen.
After lymphocytes have been stimulated by their antigen, they can acquire a memory response. The memory T
and B lymphocytes that are generated remain in the body for a long time and can respond more rapidly on repeat exposure
than naïve cells. Because of this heightened state of immune reactivity, the immune system usually can respond to
commonly encountered microorganisms so efficiently that we are unaware of the response.
Monocytes, Macrophages, and Dendritic Cells. Monocytes, tissue macrophages, and most dendritic cells arise
from a common precursor in the bone marrow. Monocytes and macrophages are key members of the mononuclear phagocytic
system. The monocytes migrate from the blood to various tissues where they mature into the major tissue phagocyte, the
macrophages. Macrophages are characterized as large cells with extensive cytoplasm and numerous vacuoles. As the general
scavenger cells of the body, macrophages can be fixed in a tissue or can be free to migrate from an organ to lymphoid tissues.
The tissue macrophages are scattered in connective tissue or clustered in organs such as the lung (i.e., alveolar macrophages),
liver (i.e., Kupffer’s cells), spleen, lymph nodes, peritoneum, central nervous system (i.e., microglial cells), and other areas.
Macrophages are activated to engulf and digest antigens that associate with their cell membrane. The initial attachment of
the microbe to the phagocyte can be aided by antibody or complement-coated microbes or by pathogen-associated molecular
pattern receptors (i.e., Toll-like receptors) that are integral to innate immune recognition.
On phagocyte membranes, the family of Toll-like receptors (so-called because they correspond in structure to a
Drosophila protein called Toll) recognizes general chemical patterns common to groups of microbes such as the
lipopolysaccharides of gram-negative bacteria or the lipoteichoic acids found in gram-positive bacteria. Once the microbe is
ingested, the cell generates digestive enzymes and toxic oxygen and nitrogen products (i.e., hydrogen peroxide or nitric
oxide) through metabolic pathways. The phagocytic killing of microorganisms helps to contain infectious agents until
adaptive immunity can be marshaled.
In addition to phagocytosis, macrophages function early in the immune response to amplify the inflammatory
response and initiate adaptive immunity. Macrophages direct these processes through the secretion of cytokines (e.g., tumor
necrosis factor [TNF], interleukin-1) that signal inflammation and activation of lymphocytes. Activated macrophages also
influence adaptive immunity as APCs that break down complex antigens into peptide fragments for association with class II
MHC molecules. Macrophages can then present these complexes to the helper T cell so that self- and non-self-recognition
and activation of the immune response can occur. Macrophages also function at the end of an immune response as effector
cells in both humoral and cell-mediated immune responses. Macrophages can remove antigen– antibody aggregates or, under
the influence of T-cell cytokines, can destroy virus-infected cells or tumor cells.
Dendritic cells share with the macrophage the important task of presenting processed antigen to T lymphocytes.
These distinctive, star-shaped cells with long extensions of their cytoplasmic membrane provide an extensive surface rich in
class II MHC molecules and other membrane molecules important for initiation of adaptive immunity. Dendritic cells are
found in most tissues where antigen enters the body and in the peripheral lymphoid tissues where they function as potent
APCs. In these different environments, dendritic cells can acquire specialized functions and appearances, as do macrophages.
Langerhans’ cells are specialized dendritic cells in the skin, whereas follicular dendritic cells are found in the lymph nodes.
Langerhans’ cells are constantly surveying the skin for antigen and can transport foreign material to a nearby lymph node.
Skin dendritic cells and macrophages also are involved in cell-mediated immune reactions of the skin such as delayed allergic
contact hypersensitivity.
B Lymphocytes. The B lymphocytes are responsible for humoral immunity. Humoral immunity provides for
elimination of bacterial invaders, neutralization of bacterial toxins, prevention of viral infection, and immediate allergic
responses.
B lymphocytes can be identified by the presence of membrane immunoglobulin that functions as the antigen
receptor, class II MHC proteins, complement receptors, and specific CD molecules. During the maturation of B cells in the
bone marrow, stem cells change into immature precursor (pre-B) cells. A rearrangement of immunoglobulin genes produces
in each cell a unique membrane receptor and secreted effector antibody (e.g., immunoglobulin M [IgM] or IgD). This stage
of maturation is programmed into the B cells and does not require antigen; it is an antigen-independent process. The various
stages of maturation can be defined by the presence of a partial or complete immunoglobulin receptor and the type of CD
molecules. The mature B cell leaves the bone marrow, enters the circulation, and migrates to the various peripheral lymphoid
tissues, where it is stimulated to respond to a specific antigen.
The commitment of a B-cell line to a specific antigen is evident by the expression of the membrane immunoglobulin
antigen receptor molecule. B cells that encounter antigen complementary to their surface immunoglobulin receptor and
receive T-cell help undergo a series of changes that transform the B cells into antibody-secreting plasma cells or into memory
B cells . B lymphocytes also can function as APCs by ingesting the surface immunoglobulin–antigen complex, processing
the antigen into small peptides, and presenting the peptide, now complexed to the class II MHC molecules, at its cell
membrane. The antigen peptide–class II MHC complex is recognized through cell-to-cell contact and stimulates the helper
T cells to secrete various cytokines. These cytokines trigger the multiplication and maturation of antigen-activated B cells.
The activated B cell divides and undergoes terminal maturation into a plasma cell, which can produce thousands of
antibody molecules per second. The antibodies are released into the blood and lymph, where they bind and remove their
unique antigen with the help of other immune effector cells and molecules. Longer-lived memory B cells are generated and
distributed into the peripheral tissues in preparation for subsequent antigen exposure.
Immunoglobulins. Antibodies comprise a class of proteins called immunoglobulins. The immunoglobulins have
been divided into five classes: IgG, IgA, IgM, IgD, and IgE, each with a different role in the immune defense strategy.
Immunoglobulins have a characteristic four-polypeptide structure consisting of at least two identical antigen-binding sites.
Each immunoglobulin is composed of two identical light (L) chains and two identical heavy (H) chains to form a Y-shaped
molecule.
The two forked ends of the immunoglobulin molecule bind antigen and are called Fab (i.e., antigen-binding)
fragments, and the tail of the molecule, which is called the Fc fragment, determines the biologic properties that are
characteristic of a particular class of immunoglobulins. The amino acid sequence of the heavy and light chains shows constant
(C) regions and variable (V) regions. The constant regions have sequences of amino acids that vary little among the antibodies
of a particular class of immunoglobulin. The constant regions allow separation of immunoglobulins into classes (e.g., IgM,
IgG) and allow each class of antibody to interact with certain effector cells and molecules. For example, IgG can tag an
antigen for recognition and destruction by phagocytes. The variable regions contain the antigen-binding sites of the molecule.
The wide variation in the amino acid sequence of the variable regions seen from antibody to antibody allows this region to
recognize its complementary epitope. A unique amino acid sequence in this region determines a distinctive three-dimensional
pocket that is complementary to the antigen, allowing recognition and binding. Each B-cell clone produces antibody with
one specific antigen-binding variable region or domain. During the course of the immune response, class switching (e.g.,
from IgM to IgG) can occur, causing the B-cell clone to produce one of the following antibody types.
IgG (gamma globulin) is the most abundant of the circulating immunoglobulins. It is present in body fluids and
readily enters the tissues. IgG is the only immunoglobulin that crosses the placenta and can transfer immunity from the
mother to the fetus. This class of immunoglobulin protects against bacteria, toxins, and viruses in body fluids and activates
the complement system. There are four subclasses of IgG (i.e., IgG1, IgG2, IgG3, and IgG4) that have some restrictions in
their response to certain types of antigens. For example, IgG2 appears to be responsive to bacteria that are encapsulated with
a polysaccharide layer, such as Streptococcus pneumoniae, Haemophilus influenzae, and Neisseria meningitidis.
IgA, a secretory immunoglobulin, is found in saliva, tears, colostrum (i.e., first milk of a nursing mother), and
bronchial, gastrointestinal, prostatic, and vaginal secretions. This dimeric secretory immunoglobulin is considered a primary
defense against local infections in mucosal tissues. IgA prevents the attachment of viruses and bacteria to epithelial cells.
IgM is a macromolecule that forms a polymer of five basic immunoglobulin units. It cannot cross the placenta and
does not transfer maternal immunity. It is the first circulating immunoglobulin to appear in response to an antigen and is the
first antibody type made by a newborn. This is diagnostically useful because the presence of IgM suggests a current infection
in the infant by a specific pathogen. The identification of newborn IgM rather than maternally transferred IgG to the specific
pathogen is indicative of an in utero or newborn infection.
IgD is found primarily on the cell membranes of B lymphocytes. It serves as an antigen receptor for initiating the
differentiation of B cells.
IgE is involved in inflammation, allergic responses, and combating parasitic infections. It binds to mast cells and
basophils. The binding of antigen to mast cell– or basophil-bound IgE triggers these cells to release histamine and other
mediators important in inflammation and allergies.
T Lymphocyte. T lymphocytes function in the activation of other T cells and B cells, in the control of intracellular
viral infections, in the rejection of foreign tissue grafts, and in delayed hypersensitivity reactions. Collectively, these immune
responses are called cell-mediated, or cellular, immunity.
Besides the ability to respond to cell-associated antigens, the T cell is integral to immunity because it regulates self-
recognition and amplifies the response of B and T lymphocytes.
T lymphocytes arise from bone marrow stem cells, but unlike B cells, pre-T cells migrate to the thymus for their
maturation. There, the immature T lymphocytes undergo rearrangement of the genes needed for expression of a unique T-
cell antigen receptor similar to but distinct from the B-cell receptor. The T-cell receptor (TCR) is composed of two
polypeptides that fold to form a groove that recognizes processed antigen peptide–MHC complexes. The TCR–antigen–
MHC complex is further stabilized by the CD4+ molecule on the helper T cell or by the CD8+ molecules on the cytotoxic T
cells. The TCR is associated with other surface molecules known as the CD3 complex that aid cell signaling. Maturation of
subpopulations of T cells (i.e., CD4+ and CD8+) also occurs in the thymus. Mature T cells migrate to the peripheral lymphoid
tissues and, on encountering antigen, multiply and differentiate into memory T cells and various effector T cells.
Helper T Cells. The CD4+ helper T cell (TH) serves as a master regulator for the immune system. Activation of
helper T cells depends on the recognition of antigen in association with class II MHC molecules. Once activated, the
cytokines they secrete will influence the function of nearly all other cells of the immune system. These cytokines activate
and regulate B cells, cytotoxic T lymphocytes, natural killer (NK) cells, macrophages, and other immune cells. The activated
helper T cell can differentiate into distinct subpopulations of helper T cells (i.e., TH1 or TH2) based on the cytokines secreted
by the APC at the site of activation. The cytokine interleukin-12 (IL-12) produced by macrophages and dendritic cells directs
the maturation of helper T cells toward TH1 cells, whereas IL-4 produced by mast cells and T cells induce differentiation
toward TH2 cells. The distinct pattern of cytokine secreted by mature.
TH1 and TH2 cells determine whether a humoral or cell-mediated response will occur. Activated TH1 cells
characteristically produce the cytokines IL-2 and interferon-γ (IFN-γ), whereas TH2 cells produce IL-4 and IL-5. In most
immune responses, a balanced response of TH 1 and TH2 cells occurs; however, extensive immunization can skew the
response to one or the other subset. For example, the extensive exposure to an allergen in atopic individuals has been shown
to shift the naïve helper T cell toward a TH2 response with the production of the cytokines that influence IgE production and
mast cell priming. An appreciation of these processes has led to clinical research suggesting that redirection of an allergic
TH2 response to a non-allergic TH1 response can occur in atopic individuals through modified immunization protocols.
T Cytotoxic Cells. Activated CD8+ cytotoxic T (Tc) cells become cytotoxic T lymphocytes (CTLs) after recognition
of class I MHC–antigen complexes on target cell surfaces, such as body cells infected by viruses or transformed by cancer.
The recognition of class I MHC–antigen complexes on infected target cells ensures that neighboring uninfected host cells,
which express class I MHC molecules alone or with self-peptide, are not indiscriminately destroyed. The CD8+ cytotoxic T
lymphocytes destroy target cells by releasing cytolytic enzymes, toxic cytokines, and pore-forming molecules (i.e., perforins)
or through programmed cell death of the target cell through triggering membrane molecules and intracellular apoptosis.
Apoptosis is a conserved cell process for the controlled elimination of excessive, dangerous, or damaged cells. In addition,
the perforin proteins can produce pores in the target cell membrane, allowing entry of toxic molecules and loss of cell
constituents. The CD8+ T cells are especially important in controlling replicating viruses and intracellular bacteria because
antibody cannot readily penetrate the membrane of living cells.
Cell-mediated immunity involves both CD4+ and CD8+ T lymphocytes. Activated CD4+ helper T cells release
various cytokines (i.e., IFN-γ) that recruit and activate other lymphocytes, macrophages, and inflammatory cells. Cytokines
(e.g., IL-8) can induce positive migration or chemotaxis of several types of inflammatory cells, including macrophages,
neutrophils, and basophils. Activation of macrophages ensures enhanced phagocytic, metabolic, and enzymatic potential,
resulting in more efficient destruction of infected cells. This type of defense is important against intracellular pathogens such
as Mycobacterium species and Listeria monocytogenes.
A similar sequence of T-cell and macrophage activation, but with sustained inflammation, is elicited in delayed
hypersensitivity reactions. Contact dermatitis due to a poison ivy reaction or dye sensitivity is an example of delayed or cell-
mediated hypersensitivity caused by hapten–carrier complexes.
Natural Killer Cells. Natural killer cells are lymphocytes that are functionally and phenotypically distinct from T
cells, B cells, and monocyte-macrophages. The NK cell is an effector cell important in innate immunity that can kill tumor
cells, virus-infected cells, or intracellular microbes. These cells are called natural killer cells because, unlike cytotoxic T
cells, they do not need to recognize a specific antigen before being activated. Both NK cells and cytotoxic T cells kill after
contact with a target cell. The NK cell is programmed to kill foreign cells automatically, in contrast to the CD8+ T cell,
which needs to be activated to become cytotoxic. However, programmed killing is inhibited in the NK cell if its cell
membrane receptors contact MHC self molecules on normal host cells.
NK cells appear as large, granular lymphocytes with an indented nucleus and abundant, pale cytoplasm containing
red granules. These cells characteristically express CD16 and CD94 cell surface molecules but lack the typical T-cell markers
(i.e., TCR, CD4). The mechanism of NK cytotoxicity is similar to T-cell cytotoxicity in that it depends on production of
pore-forming proteins (i.e., NK perforins), enzymes, and toxic cytokines. NK cell activity can be enhanced in vitro on
exposure to IL-2, a phenomenon called lymphokine-activated killer activity. NK cells also participate in antibody-dependent
cellular cytotoxicity, a mechanism by which a cytotoxic effector cell can kill an antibody-coated target cell. The role of NK
cells is believed to be one of immune surveillance for cancerous or virus-infected cells.
Cellular Receptors for Microbes, Products of Damaged
Cells that participate in innate immunity are capable of recognizing certain microbial components that are shared
among related microbes and are often essential for infectivity (and thus cannot be mutated to allow the microbes to evade
the defense mechanisms). These microbial structures are called pathogen-associated molecular patterns. Leukocytes also
recognize molecules released by injured and necrotic cells, which are called damage-associated molecular patterns.
Collectively, the cellular receptors that recognize these molecules are often called pattern recognition receptors.Pattern
recognition receptors are located in all the cellular compartments where microbes may be present:plasma membrane
receptors detect extracellular microbes, endosomal receptors detect ingested microbes, and cytosolic receptors detect
microbes in the cytoplasm. Several classes of these receptors have been identified.
Cellular receptors for microbes and products of cell injury
Phagocytes, dendritic cells, and many types of epithelial cells express different classes of receptors that sense the
presence of microbes and dead cells. Toll-like receptors (TLRs) located in different cellular compartments, as well as other
cytoplasmic and plasma membrane receptors, recognize products of different classes of microbes. The four major classes of
innate immune receptors are TLRs, NOD-like receptors in the cytosol (NLRs), C-type lectin receptors (CLRs), and RIG-like
receptors for viral nucleic acids (RLRs).
Complement System. The complement system is a primary effector system forboth innate and adaptive humoral
immune responses. Theactivation of this system results in enhanced inflammatoryresponses, lysis of foreign cells, and
increased phagocytosis.The complement system, like the blood coagulation system,consists of a group of proteins that are
present in the circulationas functionally inactive precursors. Theseproteins, mainly proteolytic enzymes, make up 10% to15%
of the plasma protein fraction. For a complement reactionto occur, the complement components must be activatedin the
proper sequence. Uncontrolled activation ofthe complement system is prevented by inhibitor proteins and the instability of
the activated complement proteins at each step of the process.
There are three parallel but independent mechanismfor recognizing microorganisms that result in the activation of
the complement system: the classic, the alternate, and the lectin-mediated pathways. All three pathways of activation generate
a series of enzymatic reactions that proteolytically cleave successive complement proteins in the pathway. The consequence
is the deposition of some complement protein fragments on the pathogen surface, thereby producing tags for better
recognition by the receptors on phagocytic cells. Other complement fragments are released into the tissue fluids to stimulate
further the inflammatory response.
The classic pathway of complement activation is initiated by antibody bound to epitopes on the surface of microbes
or through soluble immune complexes. The alternate and the lectin pathways do not use antibodies and are part of the innate
immune defenses. The alternate pathway of complement activationis initiated by the interaction of complement proteins (i.e.,
C3b) with certain polysaccharide molecules characteristic of bacterial surfaces. The lectin-mediated pathway is initiated
following the binding of a mannose-binding protein to mannose-containing molecules commonly present on the surface of
bacteria and yeast. The activation of the three pathways produces similar effects on C3 and subsequent complement proteins.
The classic pathway of complement activation was the first discovered and is the best studied. The major proteins
of the classic system are designated by a numbering system from C1 to C9. The classic pathway is triggered when
complement-fixing antibodies, such as IgG or IgM, bind to antigens. The immune complexes with complement trigger a
series of enzyme reactions that act in a cascade fashion to generate modified or split complement proteins (e.g., C3b, C3a,
and C5a). C3 has a central role in the complement pathways because it is integral to all three pathways. The triggering of C3
initiates several mechanisms for microbial destruction. One result of activation of C3 is the formation of the membrane attack
complex formed by C5 to C9. Several structurally modulated complement proteins bind to form pores in the membrane of
foreign cells that lead to eventual cell lysis.
The alternate and lectin pathways are activated by microbial surface molecules and substitute other molecules for
the proteins in the first two steps of the classic complement pathway. The alternate pathway uses proteins B, D, and P for
activation, whereas the lectin pathway uses mannose-binding protein and accessory proteins. Both pathways require the
presence of C3b and subsequent complementproteins to generate biologic effects similar to those of theclassic complement
pathway.
Whatever the mechanism of activation of the complement system, the effects of complement fixation and activation
range from cell lysis to direct mediation of the inflammatory process. First, complement has been shown to mediate the lytic
destruction of many kinds of cells, including red blood cells, platelets, and bacteria. All complement pathways may induce
cytolysis.
Second, a major biologic function of complement activation is opsonization—the coating of antigen–antibody
complexes with complement proteins such that antigens are engulfed and cleared more efficiently by macrophages.
Third, chemotactic complement products (C3a and C5a) can trigger an influx of leukocytes. These white blood cells
remain fixed in the area of complement activation through cell receptor attachment to specific sites on C3b and C4b
molecules. Fourth, production of anaphylatoxin(C3a and C5a) can activate mast cells and basophils to release biologically
active mediators (e.g., histamine) thatproduce contraction of smooth muscle, increased vascular permeability, and edema.
Major Histocompatibility Complex Molecules.An essential feature of adaptive or specific immunity is theability
to discriminate between the body’s own moleculesand foreign antigens. Key recognition molecules essentialfor
distinguishing self from nonself are the cell surface MHCmolecules. These proteins, which in humans are coded byclosely
linked genes on chromosome 6, were first identifiedbecause of their role in organ and tissue transplantation.
When cells are transplanted between individuals who are not identical for their MHC molecules, the immune system
produces a vigorous immune response leading to rejection of the transferred cells or organs. MHC molecules did not evolve
to reject transplanted tissues, a situation not encountered in nature. Rather, these molecules are essential for correct cell-to-
cell interactions among immune and body cells.
The MHC molecules involved in self-recognition and cell-to-cell communication fall into two classes, class I and
class II. Class I MHC (MHC-1) molecules are cell surface glycoproteins that interact with the antigen receptor and the CD8
molecule on cytotoxic T lymphocytes. Class I MHC molecules are found on nearly all nucleated cells in the body and are
capable of alerting the immune system of any cell changes due to viruses, intracellular bacteria, or cancer.The class I MHC
molecule contains a groove that accommodates a peptide fragment of antigen. Cytotoxic T cells can become activated only
when they are presented with the foreign antigen peptide associated with the class I MHC molecule. Antigen peptides
associate with class I molecules in cells that are infected by intracellular pathogens such as a virus. As the virus multiplies,
small peptides from degraded virus proteins associate with class I MHC molecules and are then transported to the infected
cell membrane. This complex communicates to the cytotoxic T cell that the cell must be destroyed for the overall survival
of the host. Class II MHC molecules, which are found primarily on APCs such as macrophages, dendritic cells, and B
lymphocytes, communicate with the antigen receptor and CD4 molecule on helper T lymphocytes.
Class II MHC molecules also have a groove or cleft that binds a fragment of antigen from pathogens that have been
engulfed and digested during the process of phagocytosis. The engulfed pathogen is degraded into peptide in cytoplasmic
vesicles and then complexed with class II MHC molecules. Helper T cells recognize these complexes on the surface of APCs
and then become activated. These triggered helper T cells multiply quickly and direct other immune cells to respond to the
invading pathogen through thesecretion of cytokines. A third group of genes located on the same chromosome near the class
I and class II MHC genes encode other proteins involved in the immune response. Complement and cytokines important for
signaling an immune response are examples of the third class of molecules. These secreted molecules are structurally and
functionally unrelated to the class I and class II MHC molecules.
Each individual has a unique collection of MHC proteins, and a variety of MHC molecules can exist in a population.
Thus, MHC molecules are both polygenic and polymorphic. The MHC genes are the most polymorphic genes known.
Because of the number of MHC genes and the possibility of several alleles for each gene, it is almost impossible for any two
individuals to be identical, exceptif they are identical twins. In contrast to the receptors on T and B lymphocytes that bind a
unique antigen molecule, each MHC protein can bind a broad spectrum of antigen peptides. The antigen fragments bound to
MHC molecules then allow for proper recognition of self and nonself by immune cells, and a subsequent appropriate immune
response results.
Human MHC proteins are called human leukocyte antigens (HLA) because they were first detected on white
bloodcells. Because these molecules play a role in transplantrejection and are detected by immunologic tests, theyare
commonly called antigens. More recently, analysis ofthe genes for the HLA molecules has ensured a morecomplete
identification of the potential antigens presentin an individual. The classic human class I MHC moleculesare divided into
types called HLA-A, HLA-B, and HLA-C,and the class II MHC molecules are identified as HLA-DR,HLA-DP, and HLA-
DQ. The identification or typing of HLA molecules is important in tissue or organ transplantation, forensics, and paternity
evaluations. In organ or tissue transplantation, the closer the matching of HLA types, the greater is the probability of identical
antigens and the lower the chance of rejection.
Classification of allergens:
According to their origin there can be recognized:
a. Exogenous allergens –from the environment pass in the organism;
b. Endogenous allergens – substances from bodies composition; endogenic allergens
are also called – auto-allergens.
Exogenous allergens are divided in:
1. house-keeping allergens;
2. industrial allergens;
3. drugs;
4. vegetal allergens;
5. infectious allergens;
6. parasite allergens;
This classification is of value in distinguishing the manner in which the immune response causes
tissue injury and disease, and the accompanying pathologic and clinical manifestations. However, it is
now increasingly recognized that multiple mechanisms may be operative in any one hypersensitivity
disease.
• In immediate hypersensitivity (type I hypersensitivity), the injury is caused by TH2 cells, IgE
antibodies, and mast cells and other leukocytes. Mast cells release mediators that act on vessels and
smooth muscle and pro-inflammatory cytokines that recruit inflammatory cells.
• In antibody-mediated disorders (type II hypersensitivity), secreted IgG and IgM antibodies
injure cells by promoting their phagocytosis or lysis and injure tissues by inducing inflammation.
Antibodies may also interfere with cellular functions and cause disease without tissue injury.
• In immune complex–mediated disorders (type III hypersensitivity), IgG and IgM antibodies
bind antigens usually in the circulation, and the antigen-antibody complexes deposit in tissues and
induce inflammation. The leukocytes that are recruited (neutrophils and monocytes) produce tissue
damage by release of lysosomal enzymes and generation of toxic free radicals.
• In cell-mediated immune disorders (type IV hypersensitivity), sensitized T lymphocytes (TH1
and TH17 cells and cytotoxic T lymphocytes ( CTLs)) are the cause of the tissue injury. TH2 cells induce
lesions that are part of immediate hypersensitivity reactions and are not considered a form of type IV
hypersensitivity.
The majority of effector lymphocytes induced by an infectious pathogen die by apoptosis after
the microbe is eliminated, thus returning the immune system to its basal resting state, called homeostasis.
The initial activation of lymphocytes also generates long-lived memory cells, which may survive for
years after the infection. Memory cells are an expanded pool of antigen-specific lymphocytes (more
numerous than the naive cells specific for any antigen that are present before encounter with that
antigen), and that respond faster and more effectively when re-exposed to the antigen than do naïve
cells. This is why the generation of memory cells is an important goal of vaccination.
Most circulating IgG antibodies have half-lives of about 3 weeks. Some antibody-secreting
plasma cells migrate to the bone marrow and live for years, continuing to produce low levels of
antibodies.
Sensibilization is increasing of the normal reaction to the allergen (the primary contact) to
exacerbated (from repeated contact). In this context, during immediate type allergic reactions, sensitized
body is different from the non-sensitized only by the presence of allergen-specific immunoglobulins,
which are fixed on mast cells and basophils or are in free circulation.Sensitization is the period of allergy
latency, because, until the repeated contact with the same allergen, it does not manifest clinically. Only
by serological reactions can be detected specific antibodies for free circulation, or those fixed on
basophils and mast cells. This latency period will last until the body's repeated contact with the same
allergen which caused sensitization.
II.Pathochemical stage – release, activation or synthesis of chemical mediators from altered
cells or immune cells as a result of interaction between antigen and antibody.
III.Physiopathologic stage – the stage of clinical manifestation, physiopathologic effects
launched by the mediators action on specific reactive structures in the tissues of the body.
On the contrary to sensitization stage, pathochemical and physiopathologic stages have different
mechanisms for each reaction separately.
Mast cell activation leads to degranulation, with the discharge of preformed (primary) mediators
that are stored in the granules, and de novo synthesisorsecondary mediators, including lipid products
and cytokines.
Preformed mediators. Mediators contained within mast cell granules are the first to be released
and can be divided into three categories:
• Vasoactive amines. The most important mast cell-derived amine is histamine. Histamine causes
intense smooth muscle contraction, increased vascular permeability, and increased mucus secretion by
nasal, bronchial, and gastric glands.
• Enzymes. These are contained in the granule matrix and include neutral proteases (chymase,
tryptase) and several acid hydrolases. The enzymes cause tissue damage and lead to the generation of
kinins and activatedcomponents of complement (C3a) by acting on their precursor proteins.
• Proteoglycans. These include heparin, a well-known anticoagulant, and chondroitin sulfate.
The proteoglycans serve to package and store the amines in the granules.
Other mediators which are release in the result of mast cell degranulation are: chemotactic factor
for neutrophils, chemoattractant factor for eosinophils, beta-glucosaminidase. Chemoattractantfactor
for neutrophils which trigger chemotaxis of neutrophils and their accumulation at the site of injury and
chemotactic factor for eosinophils – chemotaxis of eosinophils with inflammatory infiltration of the
tissues. In the result of neutrophils and eosinophils accumulation in the tissue and their activation, there
will be release of mediators from these inflammatory cells like: arylsulphatase, phospholipase,
histaminase, cationic proteins, that will additionally harm the tissues.
Lipid mediators. The major lipid mediators are arachidonic acid–derived products (eicosanoids
system). Reactions in the mast cell membranes lead to activation of phospholipase A2, an enzyme that
converts membrane phospholipids to arachidonic acid. This is the parent compound from which
leukotrienes and prostaglandins are produced by the lipoxygenase and cyclooxygenase pathways,
respectively (see mediators of inflammation).
• Leukotrienes. Leukotrienes C4 and D4 are the most potent vasoactive and spasmogenic agents
known. On a molar basis, they are several thousand times more active than histamine in increasing
vascular permeability and causing bronchial smooth muscle contraction. Leukotriene B 4 is highly
chemotactic for neutrophils, eosinophils, and monocytes.
• Prostaglandin D2. This is the most abundant mediator produced in mast cells by the
cyclooxygenase pathway. It causes intense bronchospasm as well as increased mucus secretion.
• Platelet-activating factor (PAF). PAF is a lipid mediator produced by some mast cell
populations but it is not derived from arachidonic acid. It causes platelet aggregation, release of
histamine, bronchospasm, increased vascular permeability, and vasodilation. Its role in immediate
hypersensitivity reactions is not well established.
Figure 3. Mast cell mediators. Upon activation, mast cells release various classes of mediators that are
responsible for the immediate and late-phase reactions. PAF, Platelet-activating factor. (from Robbins and Cotran; Pathologic
basis of disease)
Cytokines. Mast cells are sources of many cytokines, which may play an important role at several
stages of immediate hypersensitivity reactions. The cytokines include: TNF, IL-1, and chemokines,
which promote leukocyte recruitment (typical of the late-phase reaction); IL-4, which amplifies the TH2
response; and numerous others. The inflammatory cells that are recruited by mast cellderived TNF and
chemokines are additional sources of cytokines and of histamine-releasing factors that cause further
mast cell degranulation.
These mediators are responsible for the manifestations of immediate hypersensitivity reactions.
Some, such as histamine and leukotrienes, are released rapidly from sensitized mast cells and are
responsible for the intenseimmediate reactions characterized by edema, mucus secretion, and smooth
muscle spasm; others, exemplified by cytokines, including chemokines, set the stage for the latephase
response by recruiting additional leukocytes. Not only do these inflammatory cells release additional
waves of mediators (including cytokines), but they also cause epithelial cell damage. Epithelial cells
themselves are not passive bystanders in this reaction; they can also produce soluble mediators, such as
chemokines.
Late-phase reaction
In the late-phase reaction, leukocytes are recruited that amplify and sustain the inflammatory
response without additional exposure to the triggering antigen. Eosinophils are often an abundant
leukocyte population in these reactions. They are recruited to sites of immediate hypersensitivity by
chemokines, such as eotaxin, and others that may be produced by epithelial cells, TH 2 cells, and mast
cells. The TH2 cytokine IL-5 is the most potent eosinophil-activating cytokine known. Upon activation,
eosinophils liberate proteolytic enzymes as well as two unique proteins called major basic protein and
eosinophil cationic protein, which damage tissues. It is now believed that the late-phase reaction is a
major cause of symptoms in some type I hypersensitivity disorders, such as allergic asthma. Therefore,
treatment of these diseases requires the use of broad-spectrum antiinflammatory drugs, such as steroids,
rather than anti-histamine drugs, which are of benefit in the immediate reaction as occurs in allergic
rhinitis (hay fever).
Susceptibility to immediate hypersensitivity reactions is genetically determined. An increased
propensity to develop immediate hypersensitivity reactions is called atopy. Atopic individuals tend to
have higher serum IgE levels and more IL-4–producing TH2 cells than does the general population. A
positive family history of allergy is found in 50% of atopic individuals.
Environmental factors are also important in the development of allergic diseases. Exposure to
environmental pollutants, which is common in industrialized societies, is an important predisposing
factor for allergy. In fact, it is known that dogs and cats diverged from humans about 95 million years
ago and chimpanzees only about 4-5 million years ago, suggesting that chimps share more genes with
us than do dogs and cats. Nevertheless, dogs and cats, who live in the same environment as humans,
develop allergies and chimps do not. This simple observation suggests that environmental factors may
be more important in the development of allergic disease than genetics. Viral infections of the airways
are important triggers for bronchial asthma, an allergic disease affecting the lungs. Bacterial skin
infections are strongly associated with atopic dermatitis. It is estimated that 20% to 30% of immediate
hypersensitivity reactions are triggered by non-antigenic stimuli such as temperature extremes and
exercise, and do not involve TH2 cells or IgE; such reactions are sometimes called non-atopic allergy
(pseudoallergy). It is believed that in these cases mast cells are abnormally sensitive to activation by
various non-immune stimuli.
Table 2. Examples of disorders caused by immediate hypersensitivity
(from Robbins and Cotran; Pathologic basis of disease)
The incidence of many allergic diseases is increasing in developed countries, and seems to be
related to a decrease in infections during early life. These observations have led to an idea, sometimes
called the hygiene hypothesis, that early childhood and even prenatal exposure to microbial antigens
educates the immune system in such a way that subsequent pathologic responses against common
environmental allergens are prevented. Thus, too much hygiene in childhood may increase allergies later
in life. This hypothesis, however, is difficult to prove, and the underlying mechanisms are not defined.
With this consideration of the basic mechanisms of type I hypersensitivity, we turn to some clinically
important examples of IgE-mediated disease. These reactions can lead to a wide spectrum of injury and
clinical manifestations
Systemic anaphylaxis
Systemic anaphylaxis is characterized by vascular shock, widespread edema, and difficulty in
breathing. It may occur in sensitized individuals in hospital settings after administration of foreign
proteins (e.g.,antisera), hormones, enzymes, polysaccharides, and drugs (e.g., the antibiotic penicillin),
or in the community setting following exposure to food allergens (e.g., peanuts, shellfish) or insect
toxins (e.g., those in bee venom). Extremely small doses of antigen may trigger anaphylaxis, for
example, the tiny amounts used in skin testing for various forms of allergies. Because of the risk of
severe allergic reactions to minute quantities of peanuts, U.S. agencies are considering a ban on peanut
snacks served in the confined quarters of commercial airplanes. Within minutes after exposure, itching,
hives, and skin erythema appear, followed shortly thereafter by a striking contraction of respiratory
bronchioles and respiratory distress. Laryngeal edema results in hoarseness and further compromises
breathing. Vomiting, abdominal cramps, diarrhea, and laryngeal obstruction follow, and the patient may
go into shock and even die within the hour.
The risk of anaphylaxis must be borne in mind when certain therapeutic agents are administered.
Although patients at risk can generally be identified by a previous history of some form of allergy, the
absence of such a history does not preclude the possibility of an anaphylactic reaction.
Inflammation
When antibodies deposit in fixed tissues, such as basement membranes and extracellular matrix,
the resultant injury is due to inflammation. The deposited antibodies activate complement by classical
pathway, generating by-products, including chemotactic agents (mainly C5a), which direct the
migration of polymorphonuclear leukocytes and monocytes, and anaphylatoxins (C3a and C5a), which
increase vascular permeability. The leukocytes are activated by engagement of their C3b and Fc
receptors. This results in the production of other substances that damage tissues (released from activated
leucocytes), such as lysosomal enzymes, including proteases capable of digesting basement membrane,
collagen, elastin, and cartilage, and reactive oxygen species (O2-, H2O2, OH-).
Finally, we can conclude that mediators of type II allergic reaction are: compliment system,
lysosomal enzymes and reactive oxygen species.
Antibody-mediated inflammation is the mechanism responsible for tissue injury in some forms
of glomerulonephritis, vascular rejection in organ grafts, and other disorders.
Cellular dysfunction
In some cases, antibodies directed against cell surface receptors impair or dysregulate function
without causing cell injury or inflammation. For example, in myasthenia gravis, antibodies reactive with
acetylcholine receptors in the motor end plates of skeletal muscles block neuromuscular transmission
and therefore cause muscle weakness. The converse (i.e., antibody-mediated stimulation of cell
function) is the basis of Graves disease. In this disorder, antibodies against the thyroid-stimulating
hormone (TSH) receptor on thyroid epithelial cells stimulate the cells, resulting in hyperthyroidism.
Fig. 6. Recognition, display of antigen and effector cells in cell mediated hypersensibility.
Dendritic cells (DCs) capture microbial antigens from epithelia and tissues and transport the antigens to lymph nodes. During
this process, the DCs mature, and express high levels of MHC II molecules and costimulators. Naive T cells recognize MHC-
associated peptide antigens displayed on DCs. The T cells are activated to proliferate and to differentiate into effector and
memory cells (CD4+ H1 or H17, CD8+), which migrate to sites of infection and serve various functions in cell-mediated
immunity. CD4+ effector T cells of the TH1 subset recognize the antigens of microbes ingested by phagocytes, and activate
the phagocytes to kill the microbes. CD4+ T cells also induce inflammation. CD8+ cytotoxic T lymphocytes (CTLs) kill
infected cells harboring microbes in the cytoplasm. Some activated T cells differentiate into long-lived memory cells. APC,
antigen-presenting cell. (from Robbins and Cotran; Pathologic basis of disease)
Naive T lymphocytes are activated by antigen and costimulators in peripheral lymphoid organs,
and proliferate and differentiate into effector cells that migrate to any site where the antigen (microbe)
is present. One of the earliest responses of CD 4+ helper T cells is secretion of the cytokine IL-2 and
expression of high-affinity receptors for IL-2. IL-2 is a growth factor that acts on these T lymphocytes
and stimulates their proliferation, leading to an increase in the number of antigen-specific lymphocytes.
Activated CD8+ lymphocytes differentiate into CTLs that kill cells harboring microbes in the cytoplasm.
By destroying the infected cells, CTLs eliminate the reservoirs of infection.
Lymphocytes bearing specific receptors are called sensitized lymphocytes and constitute the
cellular substrate of delayed allergic reactions. The sequence of processes listed here is the phenomenon
of sensitization of the body to the allergen and consist in increased reaction to allergen - from normal
(the primary contact) to exacerbated (from repeated contact). Thus, the sensitized body differs from the
non-sensitized only by the presence of sensitized T lymphocytes, which bear on the outer surface
specific receptors for allergen.
II. Pathochemical stage start up at the repeated contact of the sensitized organism with the
allergen and consists of release, activation or synthesis of chemical mediators by sensitized
lymphocytes.
III. Physiopathologic stage – clinical manifestations – physiopathologic effects launched by
mediator release in pathochemical stage.
Responses of differentiated effector T cells. Upon repeat exposure to an antigen, TH1 cells secrete
cytokines (lymphotoxin, inhibitory factor for macrophage migration, chemoattractant factors, IFN-γ).
One of the most important from this is IFN-γ, which activates the macrophages. IFN-γ-activated
macrophages (“classically activated”) are altered in several ways: their ability to phagocytose and kill
microorganisms is markedly augmented; they express more class II MHC molecules on the surface, thus
facilitating further antigen presentation; they secrete TNF, IL-1, and chemokines, which promote
inflammation; and they produce more IL-12, thereby amplifying the TH1 response. Thus, activated
macrophages serve to eliminate the offending antigen; if the activation is sustained, continued
inflammation and tissue injury result. Activated TH17 cells secrete IL-17, IL-22, chemokines, and
several other cytokines. Collectively, these cytokines recruit neutrophils and monocytes to the reaction,
thus promoting inflammation. TH17 cells also produce IL-21, which amplifies the TH17 response.
Clinical examples of CD4+ T cell–mediated inflammatory reactions. The classic example of DTH
is the tuberculin reaction, which is produced by the intracutaneous injection of purified protein
derivative (PPD, also called tuberculin), a protein-containing antigen of the tubercle bacillus. In a
previously sensitized individual, reddening and induration of the site appear in 8 to 12 hours, reach a
peak in 24 to 72 hours, and thereafter slowly subside. Morphologically, delayed-type hypersensitivity
is characterized by the accumulation of mononuclear cells, mainly CD 4+ T cells and macrophages,
around venules, producing perivascular “cuffing”. In fully developed lesions, the venules show marked
endothelial hypertrophy, reflecting cytokine-mediated endothelial activation. With certain persistent or
non-degradable antigens, such as tubercle bacilli colonizing the lungs or other tissues, the infiltrate is
dominated by macrophages over a period of 2 or 3 weeks. With sustained activation, macrophages often
undergo a morphologic transformation into epithelioid cells, large epithelium-like cells with abundant
cytoplasm. A microscopic aggregation of epithelioid cells, usually surrounded by a collar of
lymphocytes, is referred to as a granuloma. This pattern of inflammation, called granulomatous
inflammation, is typically associated with strong TH 1-cell activation and high-level production of
cytokines such as IFN-γ. It can also be caused by indigestible foreign bodies, which activate
macrophages without eliciting an adaptive immune response.
Figure 8. Granulomatous inflammation.
The events that give rise to the formation of granulomas in type IV hypersensitivity reactions, illustrating the role of TH 1
cytokines. In some granulomas (e.g., in schistosomiasis), TH2 cells contribute to the lesions. The role of TH17 cells in
granuloma formation is not known. (from Robbins and Cotran; Pathologic basis of disease)
Contact dermatitis is a common example of tissue injury resulting from DTH reactions. It may
be evoked by contact with urushiol, the antigenic component of poison ivy or poison oak, and presents
as a vesicular dermatitis. It is thought that in these reactions, the environmental chemical binds to and
structurally modifies some self-proteins and peptides derived from these modified proteins are
recognized by T cells and elicit the reaction. Chemicals may also modify HLA molecules, making them
appear foreign to T cells. The same mechanism is responsible for most drug reactions, among the most
common immunologic reactions of humans. In these reactions, the drug (often a reactive chemical) alters
self proteins, including MHC molecules, and the “neoantigens” are recognized as foreign by T cells,
leading to cytokine production and inflammation. These often manifest as skin rashes. CD 4+ T cell–
mediated inflammation is the basis of tissue injury in many organ-specific and systemic autoimmune
diseases, such as rheumatoid arthritis and multiple sclerosis, as well as diseases probably caused by
uncontrolled reactions to bacterial commensals, such as inflammatory bowel disease.
T Cell–mediated diseases
(from Robbins and Cotran; Pathologic basis of disease)
CD8+ T cell–mediated cytotoxicity. In this type of T cell–mediated reaction, CD8+ CTLs kill
antigen-expressing target cells. Tissue destruction by CTLs may be an important component of some T
cell–mediated diseases, such as type 1 diabetes. CTLs directed against cell surface histocompatibility
antigens play an important role in graft rejection, to be discussed later. They also play a role in reactions
against viruses. In a virus-infected cell, viral peptides are displayed by class I MHC molecules and the
complex is recognized by the TCR of CD8+T lymphocytes. The killing of infected cells leads to the
elimination of the infection, but in some cases it is responsible for cell damage that accompanies the
infection (e.g., in viral hepatitis). Tumor-associated antigens are also presented on the cell surface, and
CTLs are involved in the host response to transformed cells. The principal mechanism of T cell–
mediated killing of targets involves perforins and granzymes, preformed mediators contained in the
lysosome-like granules of CTLs. CTLs that recognize the target cells secrete a complex consisting of
perforin, granzymes, and other proteins which enters target cells by endocytosis. In the target cell
cytoplasm, perforin facilitates the release of the granzymes from the complex. Granzymes are proteases
that cleave and activate caspases, which induce apoptosis of the target cells. Activated CTLs also express
Fas ligand, a molecule with homology to TNF, which can bind to Fas expressed on target cells and
trigger apoptosis. CD8+T cells also produce cytokines, notably IFN-γ, and are involved in inflammatory
reactions resembling DTH, especially following virus infections and exposure to some contact
sensitizing agents.
• T Cell–mediated reactions
The critical role of T cells in transplant rejection has been documented both in humans and in
experimental animals. T cellmediated graft rejection is called cellular rejection, and it involves
destruction of graft cells by CD8+ CTLs and delayed hypersensitivity reactions triggered by activated
CD4+ helper cells. The major antigenic differences between a donor and recipient that result in
rejectionof transplants are differences in highly polymorphic HLA alleles. The recipient's T cells
recognize donor antigens from the graft (the allogeneic antigens, or alloantigens) by two pathways,
called direct and indirect.
• In the direct pathway, T cells of the transplant recipient recognize allogeneic (donor) MHC
molecules on the surface of APCs in the graft. It is believed that dendritic cells carried in the donor
organs are the most important APCs for initiating the antigraft response, because they not only express
high levels of class I andII MHC molecules but also are endowed with costimulatory molecules (e.g.,
B7-1 and B7-2). The T cells ofthe host encounter the donor dendritic cells either within the grafted organ
or after the dendritic cells migrateto the draining lymph nodes. CD 8+ T cells recognize class I MHC
molecules and differentiate into active CTLs, which can kill the graft cells by mechanisms already
discussed. CD4+ helper T cells recognize allogeneic class II molecules and proliferate and differentiate
into TH1 (and possibly TH17) effector cells.Cytokines secreted by the activated CD4+ T cells trigger a
delayed hypersensitivity reaction in the graft, resulting in increased vascular permeability and local
accumulation of mononuclear cells (lymphocytes andmacrophages), and graft injury caused by the
activated macrophages.
• In the indirect pathway of allorecognition, recipient T lymphocytes recognize MHC antigens
of the graft donor after they are presented by the recipient's own APCs. This process involves the uptake
and processing of MHC and other foreign molecules from the grafted organ by host APCs. The peptides
derivedfrom the donor tissue are presented by the host's own MHC molecules, like any other foreign
peptide. Thus,the indirect pathway is similar to the physiologic processing and presentation of other
foreign (microbial) antigens. The indirect pathway generates CD 4+ T cells that enter the graft and
recognize graftantigens being displayed by host APCs that have also entered the graft, and the result is
a delayedhypersensitivity type of reaction. However, CD 8+ CTLs that may be generated by the indirect
pathwaycannot directly recognize or kill graft cells, because these CTLs recognize graft antigens
presented by thehost's APCs. Therefore, when T cells react to a graft by the indirect pathway, the
principal mechanism ofcellular rejection may be T-cell cytokine production and delayed
hypersensitivity. It is postulated that thedirect pathway is the major pathway in acute cellular rejection,
whereas the indirect pathway is more important in chronic rejection. However, this separation is by no
means absolute.
Fig. 9.Recognition and rejection of organ allografts.
In the direct pathway, donor class I and class II MHC antigens on antigen-presenting cells in the graft (along with
costimulators, not shown) are recognized by host CD8+ cytotoxic T cells and CD4+ helper T cells, respectively. CD4+
cells proliferate and produce cytokines (e.g., IFN-γ), which induce tissue damage by a local delayed hypersensitivity
reaction. CD8+ T cells responding to graft antigens differentiate into CTLs that kill graft cells. In the indirect pathway
graft antigens are picked up, processed, and displayed by host APCs and activate CD4+ T cells, which damage the graft by
a local delayed hypersensitivity reaction and stimulate B lymphocytes to produce antibodies.
(from Robbins and Cotran; Pathologic basis of disease)
Immunologic tolerance
Immunologic tolerance is the phenomenon of unresponsiveness to an antigen induced by exposure of lymphocytes
to that antigen. Self-tolerance refers to lack of responsiveness to an individual’s own antigens, and it underlies our ability
to live in harmony with our cells and tissues. Because the antigen receptors of lymphocytes are generated by somatic
recombination of genes in a random fashion, lymphocytes with receptors capable of recognizingself-antigens are generated
constantly, and these cells have to be eliminated or inactivated as soon as they recognize self-antigens, to prevent them from
causing harm. The mechanisms of self-tolerance can be broadly classified into two groups: central tolerance and peripheral
tolerance (Fig. 6-21). Each of these is considered briefly.
Central Tolerance In this process, immature self-reactive T and B lymphocyte clones that recognize self antigens
during their maturation in the central (or generative) lymphoid organs (the thymus for T cells and the bone marrow for B
cells) are killed or rendered harmless. The mechanisms of central tolerance in T and B cells show some similarities and
differences.
• In developing T cells, random somatic gene rearrangements generate diverse TCRs. Such antigen-independent
TCR generation produces many lymphocytes that express high-affinity receptors for self-antigens. When immature
lymphocytes encounter the antigens in the thymus, many of the cells die by apoptosis. This process, called negative selection
or deletion, is responsible for eliminating self-reactive lymphocytes from the T-cell pool. A wide variety of autologous
protein antigens, including antigens thought to be restricted to peripheral tissues, are processed and presented by thymic
antigen-presenting cells in association with self MHC molecules and can, therefore, be recognized by potentially self-reactive
T cells. A protein called AIRE (autoimmune regulator) stimulates expression of some “peripheral tissue-restricted” self-
antigens in the thymus and is thus critical for deletion of immature T cells specific for these antigens. Mutations in the AIRE
gene are the cause of an autoimmune polyendocrinopathy. In the CD4+ T-cell lineage, some of the cells that see self-antigens
in the thymus do not die but develop into regulatory T cells (described later).
• When developing B cells strongly recognize self-antigens in the bone marrow, many of the cells reactivate the
machinery of antigen receptor gene rearrangement and begin to express new antigen receptors, not specific for self antigens.
This process is called receptor editing; it is estimated that a quarter to half of all B cells in the body may have undergone
receptor editing during their maturation. If receptor editing does not occur, the self-reactive cells undergo apoptosis, thus
purging potentially dangerous lymphocytes from the mature pool. Central tolerance, however, is imperfect. Not all self
antigens may be present in the thymus, and hence T cells bearing receptors for such autoantigens escape into the periphery.
There is similar “slippage” in the B-cell system. Self-reactive lymphocytes that escape negative selection can inflict tissue
injury unless they are deleted or muzzled in the peripheral tissues.
Peripheral tolerance
Several mechanisms silence potentially autoreactive T and B cells in peripheral tissues; these are best defined for T
cells. These mechanisms include the following:
• Anergy. Lymphocytes that recognize self-antigens may be rendered functionally unresponsive, a phenomenon
called anergy. We discussed earlier that activation of antigen-specific T cells requires two signals: recognition of peptide
antigen in association with self MHC molecules on the surface of APCs and a set of costimulatory signals (“second signals”)
from APCs. These second signals are provided by certain T cell-associated molecules, such as CD28, that bind to their
ligands (the costimulators B7-1 and B7-2) on APCs. If the antigen is presented to T cells without adequate levels of
costimulators, the cells become anergic. Because costimulatory molecules are not expressed or are weakly expressed on
resting dendritic cells in normal tissues, the encounter between autoreactive T cells and their specific self-antigens displayed
by these dendritic cells may lead to anergy. Several mechanisms of T-cell anergy have been demonstrated in various
experimental systems. One of these, which has clinical implications, is that T cells that recognize self-antigens receive an
inhibitory signal from receptors that are structurally homologous to CD28 but serve the opposite functions. Two of these
inhibitory receptors are CTLA-4, which (like CD28) binds to B7 molecules, and PD-1, which binds to two ligands that are
expressed on a wide variety of cells. Because CTLA-4 has higher affinity for B7 molecules than does CD28, CTLA-4 may
be preferentially engaged when the levels of B7 are low, as when APCs are presenting self-antigens. Conversely, microbial
products elicit innate immune reactions, during which B7 levels on APCs increase and the low-affinity receptor CD28 is
engaged more. Thus, the affinities of the activating and inhibitory receptors and the level of expression of B7 may determine
the outcome of T cell antigen recognition. The importance of these inhibitory mechanisms has been established by the finding
that mice in which the gene encoding CTLA-4 or PD-1 is knocked out develop autoimmune diseases. Furthermore,
polymorphisms in the CTLA4 gene are associated with some autoimmune endocrine diseases in humans. Interestingly, some
tumors and viruses may use the same pathways of immune regulation to evade immune attack. This realization has led to the
development of antibodies that block CTLA-4 and PD-1 for tumor immunotherapy—by removing the brakes on the immune
response, these antibodies promote responses against tumors. Anergy also affects mature B cells in peripheral tissues. It is
believed that if B cells encounter self-antigen in peripheral tissues, especially in the absence of specific helper T cells, the B
cells become unable to respond to subsequent antigenic stimulation and may be excluded from lymphoid follicles, resulting
in their death. B lymphocytes also express inhibitory receptors that may play a role in limiting their activation and preventing
responses to self-antigens.
• Suppression by regulatory T cells. A population of T cells called regulatory T cells functions to prevent immune
reactions against self-antigens. Regulatory T cells develop mainly in the thymus, as a result of recognition of self-antigens
(Fig. 9), but they may also be induced in peripheral lymphoid tissues. The best-defined regulatory T cells are CD4+ cells that
express high levels of CD25, the α chain of the IL-2 receptor, and a transcription factor of the forkhead family, called FOXP3.
Both IL-2 and FOXP3 are required for the development and maintenance of functional CD 4+ regulatory T cells. Mutations
in FOXP3 result in severe autoimmunity in humans and mice; in humans these mutations are the cause of a systemic
autoimmune disease called IPEX (an acronym for immune dysregulation, polyendocrinopathy, enteropathy, X-linked). In
mice knockout of the gene encoding IL-2 or the IL-2 receptor α or β chain also results in severe multi-organ autoimmunity,
because IL-2 is essential for the maintenance of regulatory T cells. Recent genome-wide association studies have revealed
that polymorphisms in the CD25 gene are associated with multiple sclerosis and other autoimmune diseases, raising the
possibility of a regulatory T-cell defect contributing to these diseases. The mechanisms by which regulatory T cells suppress
immune responses are not fully defined, but their inhibitory activity may be mediated in part by the secretion of
immunosuppressive cytokines such as IL-10 and TGF-β, which inhibit lymphocyte activation and effector functions.
Regulatory T cells also express CTLA-4, which may bind to B7 molecules on APCs and reduce their ability to activate T
cells via CD28.
Regulatory T cells may play a role in the acceptance of the fetus. Placental mammals face a unique challenge
because the developing fetus expresses paternal antigens that are foreign to the mother yet have to be tolerated. There is
emerging evidence that regulatory T cells prevent immune reactions against fetal antigens that are inherited from the father
and therefore foreign to the mother. In line with this idea, during evolution, placentation appeared simultaneously with the
ability to stably express the Foxp3 transcription factor. Experiments in mice have shown that fetal antigens induce long-lived
Foxp3+ regulatory T cells, and depletion of these cells results in fetal loss. There is great interest in determining the
contribution of regulatory T cells in human pregnancy and possible defects in these cells as the basis for recurrent
spontaneous abortions.
• Deletion by apoptosis. T cells that recognize self-antigens may receive signals that promote their death by
apoptosis. Two mechanisms of deletion of mature T cells have been proposed, based mainly on studies in mice. It is
postulated that if T cells recognize self-antigens, they may express a pro-apoptotic member of the Bcl family, called Bim,
without anti-apoptotic members of the family like Bcl-2 and Bcl-x (whose induction requires the full set of signals for
lymphocyte activation). Unopposed Bim triggers apoptosis by the mitochondrial pathway. A second mechanism of
activation-induced death of CD4+ T cells and B cells involves the Fas-Fas ligand system. Lymphocytes as well as many other
cells express the death receptor Fas (CD95), a member of the TNF-receptor family. Fas ligand (FasL), a membrane protein
that is structurally homologous to the cytokine TNF, is expressed mainly on activated T lymphocytes. The engagement of
Fas by FasL induces apoptosis of activated T cells (Chapter 2). It is postulated that if self-antigens engage antigen receptors
of self-reactive T cells, Fas and FasL are co-expressed, leading to elimination of the cells via Fas mediated apoptosis. Self-
reactive B cells may also be deleted by FasL on T cells engaging Fas on the B cells. The importance of this mechanism in
the peripheral deletion of autoreactive lymphocytes is highlighted by two mice that are natural mutants of Fas or FasL. These
mice develop an autoimmune disease somewhat resembling human SLE, associated with generalized lymphoproliferation.
In humans a similar disease is caused by mutations in the FAS gene; it is called the autoimmune lymphoproliferative
syndrome (ALPS). Some antigens are hidden (sequestered) from the immune system, because the tissues in which these
antigens are located do not communicate with the blood and lymph. As a result, self-antigens in these tissues fail to elicit
immune responses and are essentially ignored by the immune system. This is believed to be the case for the testis, eye, and
brain, all of which are called immune-privileged sites because it is difficult to induce immune responses to antigens
introduced into these sites. If the antigens of these tissues are released, for example, as a consequence of trauma or infection,
the result may be an immune response that leads to prolonged tissue inflammation and injury. This is the postulated
mechanism for post-traumatic orchitis and uveitis.
The immune system normally exists in an equilibrium in which lymphocyte activation, which is
required for defense against pathogens, is balanced by the mechanisms of tolerance, which prevent
reactions against self-antigens. The underlying cause of autoimmune diseases is the failure of tolerance,
which allows responses to develop against self-antigens. Understanding why tolerance fails in these
diseases is an important goal of immunologists.
Immune reactions against self-antigens – autoimmunity - are an important cause of certain
diseases in humans, estimated to affect at least 1% to 2% of the US population. A growing number of
diseases have been attributed to autoimmunity. It should be noted, however, that the mere presence of
autoantibodies does not indicate an autoimmune disease exists. Autoantibodies can be found in the
serum of apparently normal individuals, particularly in older age groups. Furthermore, innocuous
autoantibodies are sometimes produced after damage to tissues and may serve a physiologic role in the
removal of tissue breakdown products. How, then, does one define pathologic autoimmunity? Ideally,
at least three requirements should be met before a disorder is categorized as truly caused by
autoimmunity: (1) the presence of an immune reaction specific for some self-antigen or self-tissue; (2)
evidence that such a reaction is not secondary to tissue damage but is of primary pathogenic significance;
and (3) the absence of another well-defined cause of the disease. Similarity with experimental models
of proven autoimmunity is also often used to support this mechanism in human diseases. Disorders in
which chronic inflammation is a prominent component are sometimes grouped under immune-mediated
inflammatory diseases; these may be autoimmune, or the immune response may be directed against
normally harmless microbes such as gut commensal bacteria.
The clinical manifestations of autoimmune disorders are extremely varied. On one end are
conditions in which the immune responses are directed against a single organ or tissue, resulting in
organ-specific disease, and on the other end are diseases in which the autoimmune reactions are against
widespread antigens, resulting in systemic or generalized disease. Examples of organ-specific
autoimmune diseases are type 1 diabetes mellitus, in which the autoreactive T cells and antibodies are
specific for β cells of the pancreatic islets, and multiple sclerosis, in which autoreactive T cells react
against central nervous system myelin. The best example of systemic autoimmune disease is SLE, in
which a diversity of antibodies directed against DNA, platelets, red cells, and protein-phospholipid
complexes result in widespread lesions throughout the body. In the middle of the spectrum falls
Goodpasture syndrome, in which antibodies to basement membranes of lung and kidney induce lesions
in these organs. It is obvious that autoimmunity results from the loss of self-tolerance, and the question
arises as to how this happens.
Autoimmune diseases
(From Robbins-Cotran; Pathological basis of disease)
Environmental factors
Although environmental factors, such as infectious agents, appear to be involved in the
pathogenesis of autoimmune disorders, their precise role in initiating or perpetuating the autoreactive
response is largely unknown.
Among the proposed mechanisms involved in loss of self-tolerance are breakdown of T-cell
anergy, release of sequestered antigens, molecular mimicry, and superantigens.
• Breakdown in T-cell anergy
Anergy is a state of unresponsiveness to antigen. It involves the prolonged or irreversible
inactivation of lymphocytes, such as that induced by an encounter with self-antigens. Activation of
antigen-specific CD4+ T cells requires two signals: (1) recognition of the antigen in association with
class II MHC molecules on the surface of an antigen-presenting cell and (2) a set of costimulatory
signals provided by the antigen-presenting cell. If the second costimulatory signal is not delivered, the
T cell becomes anergic. Most normal tissues do not express the costimulatory molecules and thus are
protected from autoreactive T cells. However, this protection can be broken if the normal cells that do
not normally express the costimulatory molecules are induced to do so. Some inductions can occur after
an infection or in situations where there is tissue necrosis and local inflammation. For example,
upregulation of the costimulator molecule B7-1 has been observed in the central nervous system of
persons with multiple sclerosis, in the synovium of persons with rheumatoid arthritis, and in the skin of
persons with psoriasis.
• Release of sequestered antigens
Normally the body does not produce antibodies against self-antigens. Thus, any self-antigen that
was completely sequestered during development and then reintroduced to the immune system is likely
to be regarded as foreign. Among the sequestered tissues that could be regarded as foreign are
spermatozoa, thyroglobulin and ocular antigens such as those found in uveal tissue. Posttraumatic uveitis
and orchiditis after vasectomy may fall into this category. Changes in antigen structure or release of
hidden antigens may also account for the persistence of autoimmune disorders. Infections, and even the
initial autoimmune episode, may expose self-antigens that have been hidden from the immune system.
The result is continued activation of new lymphocytes that recognize the previously hidden epitopes.
• Molecular mimicry
Many autoimmune diseases are associated with infections, and clinical flare-ups are often
preceded by infectious processes. One proposed link between infections and autoimmunity is molecular
mimicry, in which a microbe shares an immunologic epitope with the host. In rheumatic fever and acute
glomerulonephritis, for example, a protein in the cell wall of group A, β-hemolytic streptococci has
considerable similarity with antigens in heart and kidney tissue, respectively. After infection, antibodies
directed against the microorganism cause a classic case of mistaken identity, which leads to
inflammation of the heart or kidney. Not everyone exposed to group Aβ-hemolytic streptococci has an
autoimmune reaction. The reason that only certain persons are targeted for autoimmune reactions to a
particular self-mimicry molecule may be determined by differences in HLA types. The HLA type
determines exactly which fragments of a pathogen are displayed on the cell surface for presentation to
T cells. One individual’s HLA may bind self-mimicry molecules for presentation to T cells, and
another’s HLA type may not. In the spondyloarthropathies, particularly Reiter syndrome and reactive
arthritis, there is a clear relationship between arthritis and a prior bacterial infection, combined with the
inherited HLA-B27 antigens.
Figure 11. Postulated role of infections in autoimmunity.
Infections may promote activation of self-reactive lymphocytes by inducing the expression of
costimulators (A), or microbial antigens may mimic self antigens and activate self-reactive lymphocytes
as a cross-reaction (B); (from Robbins and Cotran; Pathologic basis of disease)
• Superantigens
Superantigens are a family of related substances, including staphylococcal and streptococcal
exotoxins, that can short-circuit the normal sequence of events in an immune response, leading to
inappropriate activation of CD4+ helper T cells. Superantigens do not require processing and
presentation of antigen by APCs to induce a T-cell response. Instead, they are able to interact with a T-
cell receptor outside the normal antigenbinding site. This distinctive mode of activation, together with
the ability of superantigens to bind to a wide variety of MHC class II molecules, leads to activation of
large numbers of T cells regardless of their MHC/peptide specificity. Superantigens are involved in
several diseases, including food poisoning and toxic shock syndrome. Recently, a bacterial superantigen
was isolated that may be important in the pathogenesis of Chron disease.
IMMUNODEFICIENCY DISEASE
Immunodeficiency can be defined as an abnormality in one or more branches of the immune
system that renders a susceptible to diseases normally prevented by an intact system. Four major
categories of immune mechanisms defend the body against infectious or neoplastic disease: humoral or
antibody-mediated immunity (B lymphocytes), cell-mediated immunity (T lymphocytes), the
complement system, and phagocytosis (neutrophils and macrophages). Humoral and cell-mediated
immunodeficiencies represent disorders of adaptive or specific immunity; and complement and
phagocytic immunodeficiencies, disorders of innate or nonspecific immunity.
Immunodeficiency states can be classified as primary (congenital or inherited) or secondary
(acquired later in life). Secondary immunodeficiency can be the result of malnutrition, infection (
acquired immunodeficiency syndrome [AIDS]), neoplastic disease (lymphoma), or immunosuppressive
therapy (corticosteroids or transplant rejection medications). Regardless of the cause, primary and
secondary deficiencies can produce the same spectrum of disease. The severity and symptomatology of
the various immunodeficiencies depend on the disorder and extent of immune system involvement.
X-linked hypogammaglobulinemia
Selective IgA deficiency. Selective IgA deficiency is the most common type of immunoglobulin
deficiency, affecting 1 in 400 to 1 in 1000 persons. The syndrome is characterized by moderate to
marked reduction in levels of serum and secretory IgA. Its likely caused by a block in the pathway that
promotes terminal differentiation of mature B cells to IgA-secreting plasma cells. The occurrence of
IgA deficiency in both males and females and in members of successive generations within families
suggests autosomal inheritance with variable expressivity. The disorder has also been noted in persons
treated with certain drugs (e.g., phenytoin, sulfasalazine), suggesting that environmental factors may
trigger the disorder. Approximately two thirds of persons with selective IgA deficiency have no overt
symptoms, presumably because IgG and IgM levels are normal and compensate for the defect. At least
50% of affected children overcome the deficiency by the age of 14 years. Persons with markedly reduced
levels of IgA often experience repeated upper respiratory and gastrointestinal infections and have
increased incidence of allergic manifestations, such as asthma, and autoimmune disorders. Persons with
IgA deficiency also can develop antibodies against IgA, which can lead to severe anaphylactic reactions
when blood components containing IgA are given. Therefore, only specially washed erythrocytes from
normal donors or erythrocytes from IgA-deficient donors should be used. There is no treatment available
for selective IgA deficiency unless there is a concomitant reduction in IgG levels. Administration of IgA
immunoglobulin is of little benefit because it has a short half-life and is not secreted across the mucosa.
There also is the risk for anaphylactic reactions associated with IgA antibodies in the immunoglobulin.
Immunoglobulin G Subclass Deficiency. An IgG subclass can affect one or more of IgG subtypes,
despite normal levels or elevated serum concentrations of IgG. IgG immunoglobulins can be divided
into four subclasses (IgG1 through IgG4) based on structure and function. Most circulating IgG belongs
to the IgG1 (70%) and IgG2 (20%) subclasses. In general, antibodies directed against protein antigens
belong to the IgG1 and IgG3 subclasses, and antibodies directed against carbohydrate and polysaccharide
antigens are primarily IgG2 subclass. As a result, persons who are deficient in IgG2 subclass antibodies
can be at greater risk for development of sinusitis, otitis media, and pneumonia caused by
polysaccharide-encapsulated microorganisms such as S. pneumoniae, H. influenzae type b, and N.
meningitidis. Children with mild forms of the deficiency can be treated with prophylactic antibiotics to
prevent repeated infections. Intravenous immune globulin can be given to children with severe
manifestations of this deficiency. The use of polysaccharide vaccines conjugated to protein carriers
rather than the protein conjugated to protein carriers that would stimulate an IgG1 response can provide
protection against some of these infections.
CELL-MEDIATED (T-CELL) IMMUNODEFICIENCIES
Unlike the B-cell lineage or immunodeficiency, in which a well-defined series of differentiation
steps ultimately leads to the production of immunoglobulins, mature T lymphocytes are composed of
distinct subpopulations whose immunologic assignments are diverse. T cells can be functionally divided
into two subtypes (CD4+ helper and CD8+ cytotoxic T cells). Collectively, T lymphocytes protect against
fungal, protozoan, viral, and intracellular bacterial infections; control malignant cell proliferation; and
are responsible for coordinating the overall immune response.
DiGeorge syndrome. DiGeorge syndrome stems from an embryonic developmental defect. The
defect is thought to occur before the 12th week of gestation, when the thymus, parathyroid gland, and
parts of the head, neck, and heart are developing. The disorder affects both sexes. Because familial
occurrence is rare, it seems unlikely that the disorder is inherited. Formerly thought to be caused by a
variety of factors, including extrinsic teratogens, this defect has been traced to microdeletion of a gene
on chromosome 22 (22q11).
Infants born with this defect have partial or complete failure of development of the thymus and
parathyroid glands and have congenital defects of the head, neck, and heart. The extent of immune and
parathyroid abnormalities is highly variable, as are the other defects. Occasionally, a child has no heart
defect. In some children, the thymus is not absent but is in an abnormal location and is extremely small.
These infants can have partial DiGeorge syndrome, in which hypertrophy of the thymus occurs with
development of normal immune function. The facial disorders can include hypertelorism (i.e., increased
distance between the eyes); micrognathia (i.e., fish mouth); low-set, posteriorly angulated ears; split
uvula; and higharched palate. Urinary tract abnormalities also are common. The most frequent
presenting sign is hypocalcemia and tetany that develops in the first 24 hours of life. It is caused by the
absence of the parathyroid gland and is resistant to standard therapy.
Children who survive the immediate neonatal period may have recurrent or chronic infections
because of impaired T-cell immunity. Children also may have an absence of immunoglobulin
production, caused by a lack of helper T-cell function. For children who do require treatment, thymus
transplantation can be performed to reconstitute T-cell immunity. Bone marrow transplantation also has
been successfully used to restore normal T-cell populations. If blood transfusions are needed, as during
corrective heart surgery, special processing is required to prevent graft-versus-host disease.
Only supportive measures are available for treatment of primary disorders of the complement
system. Measures to prevent bacterial infections are important. The affected person and close contacts
should be immunized with vaccines for S. pneumoniae, H. influenzae, and N. meningitidis.
DISORDERS OF PHAGOCYTOSIS
The phagocytic system is composed primarily of polymorphonuclear leukocytes (neutrophils
and eosinophils) and mononuclear phagocytes (circulating monocytes and tissue and fixed [spleen]
macrophages). The primary purpose of phagocytic cells is to migrate to the site of infection
(chemotaxis), aggregate around the affected tissue (adherence), envelope invading microorganisms or
foreign substances (phagocytosis), and generate microbicidal substances (enzymes or byproducts of
metabolism) to kill the ingested pathogens. A defect in any of these functions or a reduction in the
absolute number of available cells can disrupt the phagocytic system. Persons with phagocytic disorders
are particularly prone to infections by bacteria and often by Candida species and filamentous fungi,
although the types of pathogens vary with different disorders. As with other alterations in immune
function, defects in phagocytosis can be primary or secondary disorders.
BIBLIOGRAPHY
1. ROBBINS and COTRAN. Pathological basis of disease, 9th edition, 2015, pag. 186-217
3. STEFAN SILBERNAGL and FLORIAN LANG. Color Atlas of Pathophysiology. 3rd edition,
2016, pag. 56-61