Cells and Cellular Activities of the Immune System: Lymphocytes and Plasma Cells

Published on 09/02/2015 by admin

Filed under Allergy and Immunology

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 6353 times

Cells and Cellular Activities of the Immune System

Lymphocytes and Plasma Cells

Lymphocytes and Plasma Cells

The adaptive immune system is comprised of the humoral and cellular systems. Each of the two arms of the adaptive immune system has fundamental mechanisms allowing the body to attack an invading pathogen. The immunologically specific cellular component of the immune system is organized around two classes of specialized cells, T lymphocytes and B lymphocytes. Lymphocytes recognize foreign antigens, directly destroy some cells, or produce antibodies as plasma cells. The total immune response involves the interaction of many different cell types and cell-mediated and antibody-mediated responses. Recent studies have shown that T cells are not just the latecomers in inflammation, but might also play a key role in the early phase of this response. T cell subsets, including NK cells, together with classic innate immune cells, contribute significantly to the development and establishment of acute and chronic inflammatory diseases.

Lymphoid and Nonlymphoid Surface Membrane Markers

Before 1979, human lymphocytes could be classified as T or B cells based on observation of these cells with electron microscopy (Fig. 4-1). T lymphocytes have a relatively smooth surface compared with the rough pattern of the B lymphocytes.

The introduction of monoclonal antibody (MAb) testing (see Chapter 2) led to the present identification of surface membrane markers on lymphocytes and other cells. In practical terms, surface markers are used to identify and enumerate various lymphocyte subsets, establish lymphocyte maturity, classify leukemias, and monitor patients on immunosuppressive therapy.

Cell surface molecules recognized by MAbs are called antigens, because antibodies can be produced against them, or markers, because they identify and discriminate between, or “mark,” different cell populations. Originally, surface markers were named according to the antibodies that reacted with them, but a uniform nomenclature system has now been adopted.

In this system, a surface marker that identifies a particular lineage or differentiation stage with a defined structure, and that can be identified with a group or cluster of MAbs, is called a member of a cluster of differentiation (CD; Fig. 4-2). Markers can be categorized as follows:

In addition to using CD classification for the identification and separation of lymphocytes, CD antigens are involved in various lymphocyte functions, usually the following:

Sites of Lymphocyte Development

In mammalian immunologic development, the precursors of lymphocytes arise from progenitor cells of the yolk sac and liver (Fig. 4-3). Later in fetal development, and throughout the life cycle, the bone marrow becomes the sole provider of undifferentiated progenitor cells, which can further develop into lymphoblasts. Continued cellular development and proliferation of lymphoid precursors occur as the cells travel to the primary and secondary lymphoid tissues.

Primary Lymphoid Tissue

In mammals, both the bone marrow (and/or fetal liver) and thymus are classified as primary or central lymphoid organs (Fig. 4-4).

Thymus

Early in embryonic development, the stroma and nonlymphoid epithelium of the thymus are derived from the third and fourth pharyngeal pouches. The characteristics of the thymus gland change with aging. Older persons are immunologically challenged because aging causes a reduction in the production of naïve T cells by the thymus. Intrinsic defects in mature T cell function, alterations in the life span of naïve T cells and in naïve or memory T cell ratios in the peripheral lymphoid tissues, occur as the result of the decline of the T cell response in older persons.

The thymus, located in the mediastinum, exercises control over the entire immune system. It is believed that the development of diversity occurs mainly in the thymus and bone marrow, although clonal expansion can occur anywhere in the peripheral lymphoid tissue.

Progenitor cells that migrate to the thymus proliferate and differentiate under the influence of the humoral factor, thymosin. These lymphocyte precursors with acquired surface membrane antigens are referred to as thymocytes.

The reticular structure of the thymus allows a significant number of lymphocytes to pass through it to become fully immunocompetent (able to function in the immune response), thymus-derived T cells. The thymus also regulates immune function by the secretion of multiple soluble hormones.

Many cells die in the thymus and apparently are phagocytized, a mechanism to eliminate lymphocyte clones reactive against self. It is estimated that approximately 97% of the cortical cells die in the thymus before becoming mature T cells. Viable cells migrate to the secondary tissues. The absence or abnormal development of the thymus results in a T lymphocyte deficiency.

Involution of the thymus is the first age-related change occurring in the human immune system. In postnatal life, the thymus is the primary organ that produces naïve T cells for the peripheral T cell pool but production of cells declines as early as 3 months of age. The thymus gradually loses up to 95% of its mass during the first 50 years of life (Fig. 4-5). The accompanying functional changes of decreased synthesis of thymic hormones and the loss of ability to differentiate immature lymphocytes are reflected in an increased number of immature lymphocytes within the thymus and as circulating peripheral blood T cells. Most changes in immune function, such as dysfunction of T and B lymphocytes, elevated levels of circulating immune complexes, increases in autoantibodies, and monoclonal gammopathies are correlated with involution of the thymus (see Chapter 27). Immune senescence may account for the increased susceptibility of older adults to infections, autoimmune disease, and neoplasms.

Secondary Lymphoid Organs

Secondary lymphoid organs provide a unique microenvironment for the initiation and development of immune responses. The secondary lymphoid tissues include lymph nodes, spleen, GALT, thoracic duct, bronchus-associated lymphoid tissue (BALT), skin-associated lymphoid tissue, and blood. Mature lymphocytes and accessory cells (e.g., antigen-presenting cells) are found throughout the body, although the relative percentages of T and B cells vary in different locations (Table 4-1).

Table 4-1

Approximate Percentage of Lymphocytes in Lymphoid Organs

Lymphoid Organ T Lymphocytes (%) B Lymphocytes (%)
Thymus 100 0
Blood 80 20
Lymph nodes 60 40
Spleen 45 55
Bone marrow 10 90

Adapted from Claman HN: The biology of the immune response, JAMA 268:2790–2796, 1992.

The highly sophisticated structure of secondary lymphoid organs allows migration and interactions between antigen-presenting cells, T and B lymphocytes, and follicular dendritic cells (FDCs) and other stromal cells. The cooperative activities of lymphoid cells within secondary organs dramatically increase the probability of interactions of rare B, T, and APCs that results in effective generation of humoral immune responses.

Tumor necrosis factor (TNF) and lymphotoxin are essential to the formation and maintenance of secondary organs. These cytokines are produced by B and T lymphocytes. Proliferation of the T and B lymphocytes in the secondary or peripheral lymphoid tissues (Fig. 4-6) is primarily dependent on antigenic stimulation.

The T lymphocytes or T cells populate the following:

The B lymphocytes or B cells multiply and populate the following:

Circulation of Lymphocytes

Mature T lymphocytes survive for several months or years, whereas the average life span of B lymphocytes is only a few days. Lymphocytes move freely between the blood and lymphoid tissues. This activity, termed lymphocyte recirculation, enables lymphocytes to come into contact with processed foreign antigens and disseminate antigen-sensitized memory cells throughout the lymphoid system. Clonal expansion may occur regionally, as in lymph nodes draining a contact allergic reaction, and then the whole body becomes susceptible to rechallenge because T cells recirculate, but generally are excluded from returning to the thymus. Research has shown that a pool of T cell clonal elements is developed by a combination of positive selection of clones able to recognize and react to foreign antigens, and negative selection (purging) of clones able to interact with self-antigens in a damaging way.

Recirculation of lymphocytes back to the blood is through the major lymphatic ducts. Lymphocytes enter the lymph node from the blood circulation via arterioles and capillaries to reach the specialized postcapillary venules. From the venule, the lymphocytes enter the node and remain in the node or pass through the node and return to the circulating blood. Lymphatic fluid, lymphocytes, and antigens from certain body sites enter the lymph node through the afferent lymphatic duct and exit the lymph node through the efferent lymphatic duct (see Fig. 4-7).

Virgin or Naïve Lymphocytes

Virgin or naïve lymphocytes are cells that have not encountered their specific antigen. These cells do express high-molecular-weight variants of leukocyte common antigen.

Memory cells are populations of long-lived T or B cells that have been stimulated by antigen. They can make a quick response to a previously encountered antigen. Memory B cells carry surface IgG as their antigen receptor; memory T cells express the CD45RO variant of the leukocyte common antigen and increased levels of cell-adhesion molecules (CAMs), chemical mediators involved in inflammatory processes throughout the body (Fig. 4-8).

Development of T Lymphocytes

Most lymphocytes (see Color Plate 8) found in the circulating blood are T cells derived from bone marrow progenitor cells that mature in the thymus gland (Table 4-2). These cells are responsible for cellular immune responses and are involved in the regulation of antibody reactions in conjunction with B lymphocytes.

Table 4-2

Lymphocyte Characteristics

Type Function(s) Phenotypic Marker Peripheral Blood (% of Total)
Helper T (Th) cells Stimulate B cell growth and differentiation (humoral immunity); macrophage activation by secreted cytokines (cell-mediated immunity) CD3+, CD4+, CD8− 50-60
Cytotoxic T (Tc) cells Lysis of virus-infected cells, tumor cells, and allografts (cell-mediated immunity); macrophage activation by secreted cytokines (cell-mediated immunity) CD3+, CD4−, CD8+ 20-25
Natural killer (NK) cells Lysis of virus-infected cells, (antibody-dependent cellular cytotoxicity) Fc receptor for IgG or cells CD16 ∼10
B cells Antibody production (humoral immunity) Fc receptors, MHC class II, CD19, CD21 10-15

image

During cellular development, T lymphocyte function–associated antigens vary in expression. Some antigens appear early in cellular development and remain on mature T cells. Others appear at an early or intermediate stage of cellular maturation and are lost before maturity.

Early Cellular Differentiation and Development

Differentiation of a lymphocyte begins in the thymus as a thymocyte. Early surface markers on thymocytes that are committed to becoming T cells include CD44 and CD25. As thymocytes develop, there is an orderly rearrangement of the genes coding for an antigen receptor.

Maturation is a complicated process that lasts for a period of 3 weeks. During this period, cells filter through the cortex to the medulla of the thymus. Thymic stromal cells include fibroblasts, macrophages, epithelial cells, and dendritic cells; all these cell types play a role in T cell development.

Double-Negative Thymocytes

Early thymocytes lacking CD4 and CD8 surface membrane markers are referred to as double-negative thymocytes. These cells proliferate in the outer cortex of the thymus under the influence of interleukin-7 (IL7). IL-7 is critical for this growth and differentiation.

Rearrangement of the genes that code for the antigen receptor, the T cell receptor (TCR), begin at this developmental stage. CD3 constitutes the main part of the T cell antigen receptor. The configuration of two of the eight chains of the receptor have variable regions that recognize specific antigens. These are coded for by selecting gene segments and deleting others.

Rearrangement of the beta (β) chain occurs first; the appearance of a functional β chain on the cell surface sends a signal to suppress any further β chain gene rearrangements. The combination of the β chain with the CD3 forms the pre– T-cell antigen receptor (TRC). Signaling by the β chain promotes the development of a CD4+ and CD8+ thymocyte.

Thymocytes that express gamma (γ ) and delta (δ) chains follow a different developmental pathway. Cells expressing gamma-delta (γδ) chains typically remain both CD4− and CD8−. These double-negative cells represent most of the population of T lymphocytes in the skin and intestinal and pulmonary epithelium.

Circulating CD3+ double-negative lymphocytes are phenotypically and functionally distinct from single-positive CD3+CD4+ and CD3+CD8+ lymphocytes and are thought to represent a distinct T cell lineage. The presence of low numbers of double-negative T cells in healthy individuals and the increase observed in association with lymphoproliferative disorders, graft-versus-host disease, and autoimmune diseases suggest a pathogenic or immunoregulatory role for this population of T lymphocytes.

Double-Positive Thymocytes

Cells with both CD4+ and CD8+, or double-positive, surface markers represent the second stage of thymocyte development. These thymocytes begin to demonstrate rearranged genes coding for the alpha (α) chain. When the CD3-αβ receptor complex (TCR) is expressed on the cell surface, a process known as positive selection permits only double-positive cells with functional TCR receptors to survive. T cells must recognize foreign antigen in association with class I or II major histocompatibility complex (MHC) molecules. Any thymocyte that is unable to recognize self-MHC dies without ever leaving the thymus gland. Functioning T lymphocytes must be able to recognize a foreign antigen along with MHC molecules. A second selection process, negative selection, takes place among the surviving double-positive T cells. Only 1% to 3% of double-positive thymocytes survive in the cortex.

Double-positive (DP) CD4CD8 Tαβ cells have been reported in normal individuals as well as in different pathologic conditions, including inflammatory diseases, viral infections and cancer, but their function remains to be elucidated. Double-negative cells may act like natural killer (NK) cells because they are capable of binding to many natural, unprocessed cell surface molecules. In addition, these cells are capable of recognizing antigens without being presented by MHC proteins. Consequently, NK cells may represent an important bridge between natural and adaptive immunity.

Later Cellular Differentiation and Development of T Lymphocytes

When mature T cells leave the thymus, their T cell receptors (TCRs) are CD4+ or CD8+. Survivors of selection exhibit only one type of marker, CD4+ or CD8+ and migrate to the medulla. These cells gain functional maturity with their entry into the peripheral blood circulation.

T cells develop into a variety of clones. Each lymphocyte displays a single type of structurally unique receptor. The repertoire of antigen receptors in the entire population of lymphocytes is extremely large and diverse. This increases the probability that an individual lymphocyte will encounter an antigen that binds to its receptor, thereby triggering activation and proliferation of the cell. This process, clonal selection, accounts for most of the basic properties of the adaptive immune system.

Antigen receptors for common pathogens need to be reinvented by every generation of cells. Because the binding sites of antigen receptors arise from random genetic mechanisms, the receptor repertoire contains binding sites that can react not only with infectious microorganisms, but also with innocuous environmental antigens and self antigens.

Helper T Lymphocytes

Helper T lymphocytes, or T-helper (Th) cells, can be assigned to one of several subsets, including the following:

These divisions are not absolute, with considerable overlap or redundancy in function among the different subsets. This classification is based on the in vitro blends of cytokines that they produce. Th1 and Th2 cells can promote the development of cytotoxic cells and are believed to develop from Th0 cells. Th1 cells interact most effectively with mononuclear phagocytes; Th2 cells release cytokines that are required for B cell differentiation (Fig. 4-9).

Characterized by high interferon-gamma (IFN-γ) production, Th1 responses promote the elimination of intracellular pathogens (Fig. 4-10, A). Characterized by IL-4 and IL-5, Th2 responses promote a different type of effector response that involves immunoglobulin E (IgE) production and eosinophils capable of eliminating larger extracellular pathogens, such as helminths (see Fig. 4-10, B). In situations of repeated pathogen exposure or persistent infection, the polarization of T cell responses serves to focus the antigen-specific response on a specific effector pathway.

The following factors can influence the terminal differentiation of lymphocytes:

A hierarchy is apparent among these factors and is determined by how they influence T cell differentiation. Certain cytokines acting directly on T cells during primary activation appear to be the most proximal or direct mediators of CD4+ T cell differentiation. The presence of IL-12 during primary T cell activation leads to strong development of Th1 responses, and IL-4 promotes Th2 development. Activation through the TCR is a requirement for initiating terminal differentiation, but the signals from the TCR appear to be phenotype-neutral.

Certain T cells carry out delayed hypersensitivity reactions. These T cells react with antigen MHC class II on APCs and create their effects mainly through cytokine production. These cells generally are of the CD4+ phenotype.

T cells can also be differentiated into two populations depending on whether they use an αβ (TCR2) or γδ (TCR1) antigen receptor. The TCR consists of a heterodimer and a number of associated polypeptides that form the CD3 complex. The dimer recognizes processed antigen associated with an MHC molecule. The CD3 complex is required for receptor expression and is involved in signal transduction. TCR1 cells constitute less than 5% of total lymphocytes but appear in greater proportions in some sites (e.g., skin, vagina). TCR1 cells appear to recognize different antigens than TCR2 cells, including carbohydrate and intact protein antigens. In addition, some TCR1 cells do not require antigen to be processed or presented by MHC molecules.

T Regulatory Lymphocytes

Treg cells are immunoregulatory Th cells that control autoimmunity in the peripheral blood through dominant tolerance. Types of Treg cells include the following:

Natural Treg cells, characterized by constitutive expression of CD25, are developed primarily in the thymus from positively selected thymocytes with a relatively high avidity for self antigens. Natural Treg cells represent approximately 5% to 10% of the total CD4+ T cell population. The signal to develop into Treg cells is thought to come from interactions between the TCR and MHC class II self-peptide complex expressed on the thymic stroma. In humans, natural Treg cells express CD4 and CD25.

Other types of Treg cells that can develop in the periphery are Tr1 and Th3 cells. Tr1 cells are CD4+ and are functionally induced by IL-10. These Treg cells, in turn, secrete IL-10 and regulate the immune system. Th3 progenitor cells are also CD4+. In vitro CD4+ cells have been shown to secrete transforming growth factor β (TGF-β). CD8+ Treg cells are less well characterized and are reportedly capable of suppressing CD4+ cells in vitro.

Cytotoxic T Lymphocytes

Cytotoxic T lymphocytes, or T cytotoxic (Tc) cells, are effector cells found in the peripheral blood that are capable of directly destroying virally infected target cells. Most Tc cells are CD8+ and recognize antigen on the target cell surface associated with MHC class I molecules (e.g., human leukocyte antigen [HLA] types A, B, and C) or MHC class I alone. This process is demonstrated by the immune response to virus-infected cells or tumor cells (Fig. 4-11).

In a primary viral infection, naïve CD8+ T cells are primed in secondary lymph nodes and consequently proliferate and differentiate into effector CD8+ T cells to eliminate virus-infected cells. After clearance of the virus, most effector CD8+ T cells contract because of apoptosis but a small number of these CD8+ T cells form a memory T cell pool.

Studies have demonstrated that human CD8+ T cells undergo a change in the expression of costimulatory molecules (e.g., CD27, CD28, and CD45RA) on their surface, according to their differentiation and maturation. Cytolytic effector molecules, perforin, and granzyme A-B, are considered to be markers for effector CD8+ T cells because they are the actual functional molecules for killing target cells.

Naïve and central memory CD8+ T cells express the membrane marker, CCR7, for homing to secondary lymph nodes, but effector memory and effector CD8+ T cells express the chemokine receptors for inflammatory cytokines, which enable the cells to migrate toward infected and inflamed sites. A unique subset of the effector CD8+ T cell population expresses CXCR1. These CXCR1 CD8+ T cells possess chemotactic activity toward the CDCR1 ligand IL-8, a potent inflammatory cytokine produced in inflamed tissues and in tissues infected with some viruses, such as human cytomegalovirus (HCMV) or influenza A. This suggests that these CXCR1+ effector CD8+ T cells immediately migrate to inflamed and infected sites to exert their effector function in the initial stage of an immune response. It is possible that effector CD8+ T cell subsets are functionally distinct populations of T lymphocytes.

In addition to destruction of virally infected, MHC class I–bearing targets, Tc cells are major effectors in allograft organ rejection. Tc cells express CD4 or CD8, depending on the MHC antigen restriction that governs their antigen recognition (i.e., class I or II antigens; Fig. 4-12).

Suppressor T lymphocytes, or T suppressor (Ts) cells, are functionally defined T cells that downregulate the actions of other T and B cells. Ts cells have no unique markers. Although antigen-specific suppression was described in 1970, and many investigators believe that Ts cells are critical in various phases of immunoregulation, peripheral tolerance, and autoimmunity, their mode of action is unclear. Many Ts cells are CD8+ and may operate through secretion of free TCRs.

Antigen Processing and Antigen Presentation to T Cells

Antigen-presenting cells (APCs) are a group of functionally defined cells capable of taking up antigens and presenting them to lymphocytes in a form that they can recognize. APCs take up antigens (e.g., dendritic cells, macrophages, B cells, even tissue cells) in various ways. Some are collected in the periphery and transported to the secondary lymphoid tissues; other APCs normally reside in lymphoid tissues and intercept antigen as it arrives. B cells recognize antigen in a native form.

There are two major pathways of antigen processing for the APC and target cell, endogenous and exogenous. The endogenous pathway processes proteins that have been internalized, processed into fragments, and reexpressed at the cell surface membrane in association with MHC molecules. In this pathway, proteins in the cytoplasm are cleaved into peptide fragments about 20 amino acids in length. These fragments are then transported into the lumen of the endoplasmic reticulum by the transporter associated with the antigen-processing complex, where the fragments encounter newly formed, heavy-chain molecules of MHC class I and their associated beta2-microglobulin (β2m) light chains. The heavy chain, light chain, and peptide form a trimeric complex, which is then transported to and expressed on the cell surface.

T cells that express the CD8+ cell surface marker recognize antigens presented by MHC class I molecules. CD8+ functions as a coreceptor in this process, binding to an invariant region of the MHC class I molecule. Pathogen clearance requires that CD8+ effector cells produce inflammatory cytokines and develop cytolytic activity against infected target cells, after which a small number of memory cells survive that rapidly regain effector function in the event of rechallenge. During this process, a relatively homogeneous pool of naïve CD8+ T cells differentiates into heterogeneous pools of effector and memory CD8+ T cells.

In the exogenous pathway, soluble proteins are taken up from the extracellular environment, generally by specialized or so-called professional APCs. The antigens are then processed in a series of intracellular acidic vesicles called endosomes. During this process, the endosomes intersect with vesicles that are transporting MHC class II molecules to the cell surface. CD4+ T cells recognize antigens that are presented by MHC class II molecules. As with CD8, the CD4 molecule functions as a coreceptor, increasing the strength of the interaction between the T cell and APC.

For both systems of antigen presentation, recognition of the antigen by the T cells is described as being MHC-restricted, a process whereby T cells recognize only antigen presented by self MHC molecules.

Antigen Recognition by T Cells

T cells are clonally restricted, so that each T cell expresses a receptor that can interact with a given peptide. Each lymphocyte makes only one type of antigen receptor and can recognize only a very limited number of antigens. Because receptors differ on each clone of cells, the entire lymphocyte population has an enormous number of different, specific antigen receptors.

The TCR of most T lymphocytes is composed of an alpha and beta polypeptide chain, with constant regions located close to the cell surface and the part that binds to the antigenic peptide of appropriate fit located away from the cell surface. The difference in structure of the distal regions of the alpha and beta chains allows the development of different clones of T cells. The TCR reacts with antigen in the context of MHC class I or II molecules on an APC (Fig. 4-13).

T cells recognize protein antigens in the form of peptide fragments presented at the cell surface by MHC I or II molecules. When the antigen-specific TCR on the T cell surface (specifically the zeta-beta chains) of the CD3 complex interacts with the appropriate peptide-MHC complex, it triggers phosphorylation of the intracellular domains of the CD3 zeta chains. Subsequently, the zeta-associated protein 70 (ZAP-70) binds to the phosphorylated zeta chains and is activated.

Simultaneous colligation of the cell marker CD4 (or CD8) with the MHC class II (or I) molecule results in the phosphorylation of particular kinases. These events stimulate the activation of at least three intracellular signaling cascades. T cell activation also requires a second costimulatory signal (e.g., interaction between marker CD28 on T cells and marker CD80 on APCs). This interaction also triggers several intracellular signaling pathways.

T Cell Activation

T cell activation requires a minimum of two signals:

The optimal combination of effector function, proliferation, and survival requires both signals. Delivery of signal 1 without costimulation, which often occurs in tumor-infiltrating lymphocytes, leads to anergy and apoptosis, which limits the antitumor response of the cells.

Activation of T cells can lead to the following:

Activated T cells frequently express activation antigens (Box 4-1). Expression of CD69 occurs within 12 hours of activation, followed by CD25 (IL-2 receptor) and CD71 (transferrin receptor) in 1 to 3 days. Alternatively, in the case of Tc cells, interaction with antigen through the specific TCR leads to destruction of target cells.

BOX 4-1   Screening for Congenital Immunodeficiencies

Components

Adapted from Associated Regional and University Pathologists (ARUP): Lymphocyte subset panel 7: congenital immunodeficiencies, 2012 (http://www.aruplab.com/guides/ug/tests/0095899.jsp).

If a cell does not receive a full set of signals, it will not divide and may even become anergic. Peripheral T cells generally exist in a resting state (G0 or G1). T cell activation is a complex reaction involving transmembrane signaling and intracellular enzyme activation steps. It is through soluble cytokines that T cell regulation influences the action of other T cells, accessory cells, and nonimmune constituents. When activated by the proper signals, T cells may carry out one or more of the following functions:

Natural Killer and K-Type Lymphocytes

A subpopulation of circulating lymphocytes (≈10%), NK and K-type lymphocytes, lack conventional antigen receptors of T or B cells. These cells are classified as effector lymphocytes that produce mediators (e.g., IL-2).

Although these cells were previously classified as null cells, MAbs demonstrate that NK and K-type cells express a variety of surface membrane markers (Table 4-3). Most of these cells lack CD3 but express CD2, CD16, CD56, CD57 and, occasionally, CD8.

Table 4-3

Natural Killer Cell Profile

Components Reference Interval (% Positive)
CD2 75-92
CD3 63-84
CD5 61-88
CD7 73-94
CD8 14-39
CD16 1-12
CD56 7-27
CD57 1-26

Data from Associated Regional and University Pathologists (ARUP): Interpretive data guide, ed 2, Salt Lake City, Utah, 1999, Associated Regional and University Pathologists, p 473.

Natural Killer Cells

Natural killer (NK) cells are essential mediators of virus immunity. Their deficiency in humans lead to uncontrolled viral replication and poor clinical outcome. MHC class I (MCH I) is essential to NK and T cell effector and surveillance functions. A total of 70% to 80% of NK cells have the appearance of large granular lymphocytes (LGLs). Up to about 75% of LGLs function as NK cells and LGLs appear to account fully for the NK activity in mixed cell populations.

NK cells destroy target cells through an extracellular nonphagocytic mechanism referred to as a cytotoxic reaction, MHC-unrestricted cytolysis. Target cells include tumor cells, some cells of the embryo, cells of the normal bone marrow and thymus, and microbial agents. Studies have suggested that a considerable number of NK cells may be present in other tissues, particularly in the lungs and liver, where they may play important roles in inflammatory reactions and in host defense, including defense against certain viruses (e.g., cytomegalovirus, hepatitis). NK cells will actively kill virally infected target cells and, if this activity is completed before the virus has time to replicate, a viral infection may be stopped.

Several cytokines affect NK cell activation and proliferation. NK cells are highly responsive to IL-2, IL-7, and IL-12. These cytokines generate high cytokine-activated killer activity in these cells. In addition, NK cells synthesize a number of cytokines involved in the modulation of hematopoiesis and immune responses and in the regulation of their own activities.

Target cell recognition and the molecular identification and analysis of the involved NK cell receptors are undergoing intensive research. These molecules are mainly classified under the family of cell adhesion molecules (CAMs). The main class of effector CAMs shown to mediate NK cell functions is the leukocyte integrins—more specifically, the β2 class of integrins.

Several NK cell surface molecules involved in target cell recognition and binding have been identified. NK cells recognize targets using several cell surface molecular receptors (e.g., CD2, CD69, NKR-P1) and a high density of the Fc receptor CD16 of IgG (FC-R III). They also receive inhibitory signals from MHC class I on potential target cells, transduced by a killer inhibitory receptor on the NK cell. CD56 may mediate interactions between effector and target cells. NK cells are able to bind and lyse antibody-coated nucleated cells through a membrane Fc receptor that can recognize part of the heavy chain of immunoglobulins. This enables NK cells to mediate antibody-dependent, cell-mediated cytotoxic (ADCC) activities. Some, if not all, of the activation of NK cells is mediated by CD16, which exerts a regulatory role in their cytolytic function. NK cells respond to cross-linking of CD16 and CD69 as follows:

Development and Differentiation of B Lymphocytes

B cells represent a small proportion of the circulating peripheral blood lymphocytes. The unfavorable image of B lymphocytes in the pathogenesis of immune disease has been associated mainly with their capacity to produce harmful antibodies after differentiation into plasma cells.

Other roles have been discovered for B lymphocytes, including an antibody-independent pathogenic role of B cells (e.g., capability to present antigen). On recognition of a specific antigen, the B cell membrane is reorganized, resulting in an aggregation of B cell receptors in an immunologic synapse that functions as a platform for internalization of the complex. Internalized antigen is degraded and subsequently exposed to the B cell surface in association with MHC complex molecules for presentation to T cells. This surface presentation of antigen, in the presence of various costimulatory molecules, elicits the assistance of T cells required to assist B cell maturation, which in turns allows B cells to drive optimal T cell activation and differentiation into memory subsets.

B cells also have the capacity to expand clonally, which allows them to become the numerically dominant APCs. Activated B cells also produce a wide range of cytokines and chemokines that modulate the maturation, migration, and function of other immune effector cells.

B Lymphocyte Subsets

B1 and B2 cells are B cell subsets. B1 cells are distinguished by the CD5 marker, appear to form a self-renewing set, respond to a number of common microbial antigens, and occasionally generate autoantibodies. B2 cells account for most of the B lymphocytes in adults. This subset generates a greater diversity of antigen receptors and responds effectively to T-dependent antigen.

B cells are derived from progenitor cells through an antigen-independent maturation process occurring in the bone marrow and GALT. Participation of B cells in the humoral immune response is accomplished by reacting to antigenic stimuli through division and differentiation into plasma cells. Plasma cells or antibody-forming cells are terminally differentiated B cells. These cells are entirely devoted to antibody production, a primary host defense against microorganisms.

The specific antibodies produced are able to bind to infected cells, free organisms bearing the antigen, and then inactivate those cells or organisms and destroy them. The condition of hyperacute rejection of transplanted organs is also mediated by B cells. In addition, antigenic stimulation prompts B cells to multiply.

Cell Surface Markers

B lymphocytes are best known to express CD19 but not CD3 surface membrane markers. During B-cell differentiation in the bone marrow, the surface molecule CD19 appears early and remains on the B cell unit until it differentiates into a plasma cell. Four proteins on the surface of mature B cells-CD19, CD21, CD81, and CD225—from the CD19 complex.

Primitive B cell precursors have δ chains in their cytoplasm and no Ig on their surface. More differentiated (but still immature) B cells have intact cytoplasmic IgM and surface IgM. Mature B cells lose their cytoplasmic IgM and add surface IgD to the surface IgM. These changes appear to occur in the absence of antigen and depend on cytokines.

In humans, there is evidence of four types of B cell surface markers:

After binding and cooperative interaction with T cells, B cells undergo transformation into plasma cells. The secreted antibody, in turn, has the same specificity as the Ig receptor on the B cell. Almost all the antibody produced by plasma cells is secreted (plasma cells have few Ig receptors), but 90% of the antibody produced by B cells is expressed as surface Ig receptors. Some antigens (e.g., lipopolysaccharides from some gram-negative organisms) can bind to the Ig receptor and also stimulate an antibody response independent of T cell cooperation (T-independent antigens). This type of response is generally of low intensity and is class-restricted to the production of IgM antibody.

B cells have surface immunoglobulin (sIg), except for very immature lymphocytes and mature plasma cells, that are normally polyclonal (i.e., kappa and lambda light chains are present on the cytoplasmic membrane of B cells). Mu and delta heavy chains are usually found with kappa or lambda chains on any one cell surface. Gamma and alpha chains are rarely found on the surface of properly prepared, normal lymphocytes.

B Cell Activation

B cells can be stimulated in their resting state to enlarge, develop synthetic machinery, divide, mature, and secrete antibody. The proper signals for this sequence depend on the type of triggers, which can be specific or nonspecific and polyclonal. Specific activation involves the antigen that is complementary to the particular Ig on the surface. Nonspecific activation occurs with B cell mitogens.

Efficient antibody production to complex protein antigens requires T cell help, which in turn develops from APCs presenting antigen to the T cell. Activated T cells secrete a variety of cytokines that together with the specific antigen, trigger the B cell to develop into an antibody-secreting cell. This process also involves class switching.

In the immune response to a foreign protein, the first antibodies to appear are of the IgM class (or isotype). As the response proceeds, other isotypes (IgG, IgA, and IgE) emerge from Ig class switching. The isotype switch has considerable clinical importance because each of the four major isotypes has specialized biologic properties. IgG is the principal class of antibody in interstitial fluids and IgA is the protective antibody of mucosal surfaces. Isotype switching requires collaboration between antibody-synthesizing B cells and helper CD4+ T cells. The B cell uses IgM molecules on its surface to capture the antigen and present the antigen to the T cell. Contact between the collaborating lymphocytes is enhanced by complementary pairs of CAMs. Some CAMs (e.g., CD4, MHC class II antigens) are constitutively expressed on the surface of T and B cells, whereas others are induced. For example, contact between B and T cells induces the T cell to express a ligand for the B cell surface molecule CD40. In turn, CD40 interacts with the newly expressed CD40 ligand on the T cell, which leads to the expression of another B cell surface molecule, B7. The latter’s partner on the surface of the T lymphocyte is CD28. These cooperative and synergistic interactions between T and B cells induce the secretion of cytokines such as IL-2 and IL-4.

Isotype switching requires two signals. The first is delivered by an interleukin and the second by the binding of CD40 to its ligand on the T cell. In the process of switching from IgM synthesis to IgE synthesis, IL-4 makes the IgE gene in the B cell accessible to the switch machinery initiated when CD40 binds to its ligand. In this process, the gene that encodes the variable region (the part of the antibody molecule that contains the antigen-binding site) moves from its position near the gene that encodes for IgM to a position near the gene that encodes for IgE.

Plasma Cell Biology

The function of plasma cells (see Color Plate 9) is the synthesis and excretion of immunoglobulins. Plasma cells are not normally found in the circulating blood but are found in the bone marrow in concentrations that do not normally exceed 2%. Plasma cells arise as the end stage of B cell differentiation into a large, activated plasma cell.

The pathway from the B lymphocyte to the antibody-synthesizing plasma cell forms when the B cell is antigenically stimulated and undergoes transformation because of the stimulation of various interleukins. The immune antibody response begins when individual B lymphocytes encounter an antigen that binds to their specific Ig surface receptors. After receiving an appropriate second signal provided by interaction with helper T cells, these antigen-binding B cells undergo transformation and proliferation to generate a clone of mature plasma cells that secretes a specific type of antibody.

An increase in plasma cells can be seen in a variety of nonmalignant disorders, such as viral disease (e.g., rubella, infectious mononucleosis), allergic conditions, chronic infections, and collagen diseases. In plasma cell dyscrasias, the plasma cells can be greatly increased or infiltrate the bone marrow completely (e.g., multiple myeloma, Waldenström’s macroglobulinemia).

Antibody molecules secreted by plasma cells consist of four chains—two light chains and two heavy chains, based on molecular weight—and can be enzymatically cleaved into Fab (antigen-binding) and Fc (crystallizable) fragments. The Fab portion binds antigen and contains the light chains and their antigenic markers (kappa, lambda), as well as heavy chains.

The Fc fragment contains the markers that distinguish the different classes of antibody and sites that will bind and activate complement and bind to Fc receptors on cells. The amino acid sequence for most of the antibody protein is constant, except for the antigen-binding portion of the molecule, which has a hypervariable region and accounts for the various antigenic specificities that the antibody is programmed to recognize.

Alterations in Lymphocyte Subsets

The normal functioning of helper cells and suppressor cells in the immune response can be reversed under certain conditions. For example, the target cell for human T cell leukemia or human immunodeficiency virus (HIV) is phenotypically a helper cell but functionally a suppressor cell. Functionally, the helper-inducer subset of cells signals B cells to generate antibodies, control production and switching of types of antibodies formed, and activate suppressor cells. The suppressor-cytotoxic lymphocytes control and inhibit antibody production by suppressing helper cells or by turning off B cell differentiation. The normal ratio of helper cells and suppressor cells (≈2:1) can be reversed under certain conditions.

Changes With Aging

Except for inconsistent values seen in extremely old adults, the total number of T cells in the peripheral blood is relatively stable throughout adult life. However, there is a change in the distribution of T cell subpopulations. A decrease in the number of suppressor cells and an increase in the helper cell population are demonstrated in older adults.

The effect of aging on the immune response is highly variable, but the ability to respond immunologically to disease is age-related. Faulty immunologic reactions (e.g., aberrant functioning of immunoregulatory cells, effector T cells, and antibody-producing B cells) may contribute to poor immunity in older adults. Functional deficits of T lymphocytes have been identified with aging, causing impairment of cell-mediated immunity. In addition, skin testing reveals decreases in the intensity of delayed hypersensitivity in older adults. The proliferative response of T lymphocytes to mitogens or antigens such as Mycobacterium tuberculosis or varicella-zoster virus is impaired.

A decrease in Th cells is the primary cause of the impaired humoral response in older adults. Although the total number of B cells and total Ig concentration remain unchanged, the serum concentration of IgM is decreased and IgA and IgG concentrations are increased.

Evaluation of Immunodeficiency Syndromes

Although more than 50 genetically determined immunodeficiency syndromes have been reported since 1952, defects in immunity were considered rare until acquired immunodeficiency syndrome (AIDS) emerged more than 30 years ago. This growing list of primary and secondary diseases now encompasses all major components of the immune system, including lymphocytes, phagocytic cells, and complement proteins.

Older children and adults with recurrent upper and lower respiratory tract infections and/or diarrhea, abscesses, sepsis, or meningitis should be evaluated for immunodeficiency. Before proceeding with laboratory testing, primary care providers need to rule out the following:

Laboratory testing can then proceed with a complete blood cell (CBC) count, including a platelet count and erythrocyte sedimentation rate (ESR). These are among the most cost-effective screening tests. If the ESR is normal, chronic bacterial infection is unlikely. If the absolute neutrophil count is normal, congenital and acquired neutropenias and severe chemotactic defects are eliminated. If the absolute lymphocyte count is normal, the patient is not likely to have a severe T cell defect. The absolute lymphocyte count is the number of lymphocytes in the total white blood cell (WBC) population (Box 4-2).

Laboratory tests to screen for more common immunodeficiencies include immunoglobulin testing, complement testing, cell-mediated immunity testing, and the neutrophil function test. Additional laboratory testing should include a general metabolic panel to assess overall general health, HIV types 1 and 2, protein electrophoresis, sweat chloride, and pneumococcal antibody IgG titers pre- and postvaccine in patients with only recurrent sinopulmonary infections. Follow-up testing based on any initially abnormal results is presented in Tables 4-4 and 4-5. If all initial test results are normal, IL-1 receptor-associated kinase-4 (IRAK-4) deficiency screening or a Toll-like receptor function assay should be performed. If abnormal results are subsequently found, the diagnosis is an innate immune deficiency.

Table 4-4

Next Steps in Laboratory Evaluation of Suspected Immunodeficiency

Abnormal Initial Laboratory Result Follow-Up Testing
Protein electrophoresis Immunofixation electrophoresis monoclonal protein detection; quantitation and characterization of IgA, IgG and IgM, and Bence Jones protein; depending on individual results, additional testing may be needed
Sweat chloride Cystic fibrosis (CFTR)—32 mutations with reflex to sequencing
Positive for HIV-1, HIV-2 HIV-1 antibody confirmation by Western blot
Pneumococcal antibodies absent after vaccination Specific antibody deficiency

Adapted from Associated Regional and University Pathologists (ARUP) Consult: Immunodeficiency evaluation for chronic infections in adults and older children testing algorithm, 2012 (http://www.arupconsult.com/Algorithms/ChronicInfections Adults.pdf).

Table 4-5

Next Steps in Evaluation of Suspected Immunodeficiency Based on Physical Findings

Physical Manifestations Follow-Up Laboratory Assays Differential Diagnosis
Recurrent severe viral or fungal infections (e.g., candidiasis, herpes) Lymphocyte subset for congenital immunodeficiencies, lymphocyte antigen and mitogen proliferation panel, proliferation panel with cytokine responses to mitogens; testing for 12 cytokines T cell deficiency, HIV, CD4 deficiency, adenosine deaminase deficiency, nucleoside phosphorylase deficiency, chronic mucocutaneous candidiasis
Recurrent severe sepsis, Neisseria spp., Streptococcus pneumoniae infections Rule out previous splenectomy and immunoglobulin abnormality; then order a total CAEI. If abnormal (low) activity, analyze C2-C5 components. Complement deficiency
Abscesses, pneumonia, recurrent respiratory infections with or without diarrhea Order quantitative IgM, IgG and IgA, IgE, CAEI, neutrophil oxidative burst assay (DHR), leukocyte adhesion deficiency panel, and myeloperoxidase stain. Low IgM– or IgG–hypogammaglobulinemia
Low complement—complement defect
Abnormal DHR—chronic granulomatous disease
Decreased CD11b/CD18—LAD-1,
Decreased CD15—LAD-2
Increased IgE—Possible hyper-IgE syndrome (Job syndrome) but must be followed by Candida-specific IgE neutrophil chemotaxis and subsequent genetic testing
Positive neutrophil antibody—autoimmune neutropenia,
Absence of myeloperoxidase—myeloperoxidase deficiency

CAEI, Complement activity enzyme immunoassay; DHR, neutrophil oxidative burst assay; LAD-1, leukocyte adhesion deficiency, type 1; LAD-2, leukocyte adhesion deficiency, type 2.

Adapted from Associated Regional and University Pathologists (ARUP) Consult: Immunodeficiency evaluation for chronic infections in adults and older children testing algorithm, 2012 (http://www.arupconsult.com/Algorithms/ChronicInfections Adults.pdf).

Cell-Mediated Immune System

Deficiencies of cell-mediated immunity are often suspected in individuals with recurrent viral, fungal, parasitic, and protozoal infections. Patients with AIDS exhibit some of the most severe manifestations of cell-mediated immunity (see Chapter 25).

One avenue of testing involves delayed hypersensitivity skin testing to determine the integrity of the patient’s cell-mediated immune response. More than 90% of normal adults will react to one of the following antigens within 48 hours after antigen exposure: Candida albicans, Trichophyton, tetanus toxoid, mumps, and streptokinase-streptodornase. Reactivity to histoplasmin or purified protein derivative (PPD) is positive in patients with active infection or previous exposure to histoplasmosis or tuberculosis, respectively; therefore these tests are not useful for the assessment of anergy.

The number of T lymphocytes, the primary effector cells in cell-mediated reactions, can be determined by several techniques. Previously, the gold standard was the E rosette technique (erythrocyte rosette formation), but the development of flow cytometry with MAbs has replaced this technique. Testing for the functionality of lymphocytes is just as important as a quantitative count of CD4+ and CD8+ cells.

The in vitro diagnostic test (IVD; see later, “Assessment of Cellular Immune Status”) is the newest approach to testing the functionality of T lymphocytes. It is important to recognize that CD4 counts do not always reflect the actual status of the patient’s immune system. ImmuKnow (Cylex, Columbia, Md) is the first U.S. Food and Drug Administration (FDA)–approved immune function test. It is widely considered to be the gold standard of immune function testing.

The QuantiFERON-CMI kit (Cellestis, Valencia, Calif) is an in vitro assay for measuring cell-mediated immune functionality. The procedure is a single-step enzyme-linked immunosorbent assay (ELISA) to determine T cell responses by measuring IFN-γ levels in plasma. This is a specific marker cytokine for a cell-mediated or inflammatory immune response (e.g., bacterial, parasitic, or viral).

Research-Based Tests

Various procedures are research-based, including a lymphocyte antigen and mitogen proliferation panel. This type of procedure measures cytokine production by mononuclear cells in response to mitogen stimulation by IL-1β types 6 and 8 and TNF-α. Another method includes flow cytometry and the enzyme-linked immunosorbent spot assay (ELISPOT). Flow cytometry can be used in conjunction with intracellular cytokine staining with 3H-thymidine to detect the T cell response to specific antigenic stimulation by multianalyte fluorescence detection. In addition, peptide–MHC complex tetramer or pentamer staining is used to quantify the number of T cells with a particular antigenic epitope based on the expression of a specific T cell receptor. A multiplex cytokine analysis (e.g., multiplex bead-based Luminex assay [Life Technologies, Grand Island, NY]) is being used to detect multiple cytokines in serum, plasma, or tissue culture supernatants.

Immunologic Disorders

A breakdown in any part of the immune mechanism can lead to disease. Disorders with an immunologic origin can involve progenitor cells, phagocytosis (see Chapter 3), T cells, B cells, or complement (see Chapter 5).

Immunologic disorders can be divided into primary processes (dysfunction in the immune organ itself) and acquired, or secondary, processes (disease or therapy causing an immune defect). A third category, diseases mediated through immune mechanisms, can also be included. Because of its complexity and contemporary importance, AIDS is discussed separately in Chapter 25. Other immunoproliferative and autoimmune disorders are discussed in Chapters 27 to 30.

Immunodeficiency disorders may be caused by defects in the quality (defects) or quantity (deficiencies) of lymphocytes and may be congenital or acquired. These conditions may be combined disorders or may involve T cells or B cells (Table 4-6).

Table 4-6

T Cell and B Cell Disorders

T Cell Disorder B Cell Disorder
Congenital
Thymic hypoplasia (DiGeorge’s syndrome) Bruton’s agammaglobulinemia
Acquired
Acquired immunodeficiency syndrome Autoimmune disorders
Hodgkin’s disease Multiple myeloma
Chronic lymphocytic leukemia  
Systemic lupus erythematosus  

image

Primary Immunodeficiency Disorders

Primary immunodeficiencies (PID) are rare genetic disorders of the innate and adaptive immune system. Classic primary immunodeficiency disorders (PIDs) are usually monogenic (mendelian) disorders affecting host defenses (Box 4-3). More than 200 clinical phenotypes of PID have been described. Over 120 different gene mutations have been identified which cause impairment in the differentiation and/or function of immune cells with different degrees of severity. Diseases associated with a primary defect in the immune response are comprised of about 40% T cell disorders, 50% B cell disorders, 6% phagocytic abnormalities, and 4% complement alterations (Fig. 4-14). The most common T cell deficiency states are those associated with a concurrent B cell abnormality. Primary immunodeficiency disorders are predominantly seen (75%) in children younger than 5 years.

Gene therapy with hematopoietic stem cells (HSC) is a therapeutic strategy for the treatment of several forms of primary immunodeficiency. Current approaches use gene transfer of the therapeutic gene into autologus HSC by retroviral vector-mediated gene transfer. This method has been successful in severe combined immunodeficiencies (SCID-1) and chronic granulomatous disease (CGD). Wiskott-Aldrich syndrome is another good candidate for gene therapy treatment.

T Cell and Combined Immunodeficiency Disorders

DiGeorge’s Syndrome

Immunologic Manifestations

Peripheral lymphoid tissue appears to be normal except for the depletion of T cells in thymus-dependent zones, such as subcortical region of the lymph nodes and perifollicular and periarteriolar lymphoid sheaths of the spleen. Lymph node paracortical areas and thymus-dependent regions of the spleen show variable degrees of depletion.

In the circulating blood, lymphopenia is generally present, although in some cases the concentration of lymphocytes is normal. However, an abnormally high CD4+/CD8+ ratio is present because of a decrease in CD8+ cells. Most patients with DiGeorge’s syndrome have a decreased percentage of cells expressing the CD3+ (mature T cell) antigen. Because patients do demonstrate lymphocytes capable of differentiating to the more mature surface markers, such as CD4+, a small rudimentary thymus is believed to be present in these patients. Lymphocytic responsiveness to antigenic and mitogenic stimulation can be absent, reduced, or normal, depending on the degree of thymic deficiency. Cell-mediated immune reactions such as delayed hypersensitivity and skin allograft rejections, however, are absent or feeble.

Serum Ig concentrations are near normal. Levels of IgA may be diminished and of IgE may be elevated. Antibody response to primary antigenic stimulation may be unimpaired.

Nezelof Syndrome (Cellular Immunodeficiency With Immunoglobulins)

Severe Combined Immunodeficiency

Cause

Severe combined immunodeficiency (SCID) is caused by the inappropriate development of progenitor cells into lymphocyte precursors. This hereditary and invariably fatal disorder in infants results from the lack of both T and B cells and the consequent inability to synthesize antibody.

Mutations in the IL-2 receptor complex, a hematopoietic growth factor, have been shown to cause X-linked SCID in humans. Two modes of inheritance are known, autosomal recessive and X-linked recessive. X-linked recessive SCID is thought to be the most common form of SCID in the United States, which accounts for the 3:1 male-to-female ratio with the disorder.

Of patients with autosomal SCID, 50% have a concomitant deficiency of adenosine deaminase (ADA), an aminohydrolase that converts adenosine to inosine. Analysis by complementary (copy) DNA (cDNA) probe has revealed that the deficiency results from a hereditable point mutation in the ADA gene. Another variant with a severe deficiency in T cell immunity but normal B cell concentrations is associated with purine nucleotide phosphorylase deficiency.

There are two main forms of defective expression of MHC antigens. In a less common form of SCID, known as bare lymphocyte syndrome, an MHC class I antigen deficiency is present. In another form of defective expression, MHC class I antigen deficiency plus the absence of class II antigens is present. Patient lymphocytes cannot be typed by standard serologic cytotoxicity tests.

Immunologic Manifestations

The thymus and other lymphoid organs are severely hypoplastic. The bone marrow is devoid of lymphoblasts, lymphocytes, and plasma cells. Lymphocytes are also absent from lymphoid tissues such as the spleen, tonsils, appendix, and intestinal tract. Variable hypogammaglobulinemia with decreased serum IgM and IgA levels and poor to absent antibody production are representative features. Moderate lymphocytopenia is detectable early in infancy. T cell functions are decreased. The circulating blood contains no CD4+, CD8+, or CD3+ cells. The percentage of B cells is usually normal.

Patients with the X-linked form of SCID usually appear similar to those with the autosomal recessive form, except that they tend to have an increased percentage of B cells. However, the defect affects B-lineage cells as well as T-lineage cells.

B Cell and Antibody Deficiency Disorders

Because the primary function of B cells is to produce antibody, the major clinical manifestation of a B cell deficiency is an increased susceptibility to severe bacterial infections. Selective IgA deficiency is the most common B cell disorder, affecting 1 in 400 to 800 persons. Because IgA is the primary immunoglobulin in secretions, a deficiency contributes to pulmonary infections, gastrointestinal (GI) disorders, and allergic respiratory disorders. Most cases (50% of reported cases are associated with Ig deficiencies) are autoimmune in nature, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), thyroiditis, and pernicious anemia.

Bruton’s X-Linked Agammaglobulinemia

Signs and Symptoms

X-linked agammaglobulinemia occurs primarily in young boys, but scattered cases have been identified in girls. Manifestations begin in the first or second year of life. Hypersusceptibility to infection does not develop until 9 to 12 months after birth because of passive protection by residual maternal immunoglobulin. Thereafter, patients repeatedly acquire infections with high-grade extracellular pyogenic organisms such as streptococci. This disorder is characterized by sinopulmonary and central nervous system (CNS) infectious episodes and severe septicemia, but patients are not abnormally susceptible to common viral infections (excluding fulminant hepatitis), enterococci, or most gram-negative organisms. Chronic fungal infections are not usually present.

An autoimmune phenomenon, especially a juvenile RA type of disease, has also been associated with X-linked agammaglobulinemia. In addition, patients are highly vulnerable to a malignant form of dermatomyositis that eventually involves destructive T cell infiltration surrounding the small vessels of the CNS. In addition to infections and connective tissue disorders, agammaglobulinemic patients also have hemolytic anemia, drug eruptions, atopic eczema, allergic rhinitis, and asthma.

Immunologic Manifestations

The diagnosis of X-linked agammaglobulinemia is suspected if serum concentrations of IgG, IgA, and IgM are notably below the appropriate level for the patient’s age. Tests for natural antibodies to blood group substances and for antibodies to antigens given during standard courses of immunization (e.g., diphtheria) are useful in distinguishing this disorder from transient hypogammaglobulinemia of infancy (see later).

B cells are almost absent from bone marrow and lymphoid tissues. A deficiency or absence of peripheral B lymphocytes is usually noted. If present, B cells are unresponsive to T cells and incapable of antibody synthesis or secretion. Surface immunoglobulins are absent. However, patients have normal numbers of CD3+ and CD8+ cells and many have normal CD4+ cells. Male children possess normal T cell function; therefore, homograft rejection mechanisms are intact and delayed-hypersensitivity reaction for both tuberculin and skin contact types can be elicited.

Common Variable Immunodeficiency

Immunologic Manifestations

Both the decreased concentration of immunoglobulins and near absence of serum and secretory IgA are thought to represent the most common and well-defined type of PID. The pattern of inheritance suggests that an autosomal function of antibodies is usually compromised. The number of B cells is typically normal or mildly depressed. Despite a normal number of circulating Ig-bearing B lymphocytes and the presence of lymphoid cortical follicles, blood lymphocytes do not differentiate into Ig-producing cells. In most patients the defect appears to be intrinsic to the B cell. The primary defect in Ig synthesis may be caused by the absence or dysfunction of CD4+ cells or by increased CD8+ supressor cell activity. Therefore, cellular immunity and Ig production are impaired by the interaction between helper and suppressor T cell subsets. Lymph nodes lack plasma cells, but may show striking follicular hyperplasia.

The total IgG level may be normal, but a subclass (usually IgG2 or IgG3) is deficient. Both IgA and IgM may be detectable, but IgM levels may be elevated. In addition, some patients may have thymoma and refractory anemia.

Immunoglobulin Subclass Deficiencies

Some patients have deficiencies of one or more subclasses of IgG despite a normal total IgG serum concentration. Most of those with absent or very low concentrations of IgG2 have been patients with selective IgA deficiency.

Selective Immunoglobulin A Deficiency

Immunodeficiency With Elevated Immunoglobulin M (Hyper-IgM)

X-Linked Lymphoproliferative Disease (Duncan’s Disease)

Signs and Symptoms

The disease is characterized by an inadequate immune reaction to infection with Epstein-Barr virus (EBV). Infected patients are apparently healthy until they experience infectious mononucleosis (see Chapter 22). Two thirds of more than 100 patients studied died of overwhelming EBV-induced B cell proliferation during mononucleosis. Most patients surviving the primary infection developed hypogammaglobulinemia and/or B cell lymphomas.

Partial Combined Immunodeficiency Disorders

Wiskott-Aldrich Syndrome (Immunodeficiency With Thrombocytopenia and Eczema)

Hereditary Ataxia-Telangiectasia

T Cell Activation Defects

Some patients with defective activation of T cells have experienced the following:

These conditions are characterized by the presence of T cells that appear phenotypically normal but fail to proliferate or produce cytokines in response to stimulation with mitogens, antigens, or other signals delivered to the T cell antigen receptor. Patients’ symptoms are similar to those of other T cell–deficient individuals; some patients with severe T cell activation defects may clinically resemble patients with SCID.

Other Primary Immunodeficiencies

In addition to hereditary or congenital disorders of lymphocytes, several PIDs involve the complement system and phagocytic cells.

Complement Deficiency

Deficiencies in all of the components of the complement system have been described (see Chapter 5). These deficiencies are genetic in origin. Unusual susceptibility to infection is characteristic of some of these components, particularly deficiencies involving C3, C5, C6, and C7 (Table 4-7).

Table 4-7

Complement Deficiencies

Deficient Component Common Types of Infections
C1 (r/q) Gram-positive, mainly respiratory
C2 Gram-positive, recurrent respiratory; meningitis, sepsis, tuberculosis
C3 Gram-positive, recurrent
C4 Gram-positive; sepsis, meningitis
C5 Meningitis (Neisseria meningitidis), disseminated gonococcal infection
C6 Meningitis (N. meningitidis), disseminated gonococcal infection
C7 Meningitis (N. meningitidis)
C8 Meningitis (N. meningitidis), disseminated gonococcal infection
C9 Meningitis (N. meningitidis)

A functional deficiency of polymorphonuclear neutrophil (PMN) leukocytes is chronic granulomatous disease (CGD; see Chapter 3). This fatal syndrome usually begins with the onset of symptoms during the first year of life.

Secondary Immunodeficiency Disorders

A secondary immunodeficiency can result from a disease process that causes a defect in normal immune function, which leads to a temporary or permanent impairment of one or multiple components of immunity in the host (Box 4-4). Patients with secondary immunodeficiencies, which are much more common than primary deficiencies, have an increased susceptibility to infections, as seen in the PIDs.

Immunosuppressive agents and burns are major causes of secondary immunodeficiencies. In varying degrees, immunosuppressive agents have been demonstrated to affect every component of the immune response. In burn patients, septicemia is a common complication in those who survive the initial period of hemodynamic shock. The mechanism that seems most critical in thermal injury is disruption of the skin; however, interference with phagocytosis and deficiencies of serum Ig and complement levels have also been observed.

Immune-Mediated Disease

The immune system is normally efficient in eliminating foreign antigens. The nature of the antigen or the genetic makeup of the host, however, can cause alterations of the immune response that can be injurious and lead to immune-mediated disease (Table 4-8). In these disorders, the immune response is normal but the reactivity is heightened, prolonged, or inappropriate.

Table 4-8

Immune-Mediated Disease

Type Cause, Disease
Allergic hypersensitivity Foods, drugs, aeroallergens (dust, pollens, molds)
Contact hypersensitivity Poison ivy, nickel, cosmetics
Transfusion Reactions
Autoimmune disease Systemic lupus erythematosus, rheumatoid arthritis, vasculitis syndromes, hemolytic anemia, idiopathic thrombocytopenia, pernicious anemia, Goodpasture’s syndrome, myasthenia gravis, Graves’ disease

image

A major concern is allergic reactions, characterized by an immediate response on exposure to an offending antigen and the release of mediators (e.g., histamine, leukotrienes, prostaglandins) capable of initiating signs and symptoms (see Chapter 26). Although allergic reactions are associated with IgE, not all allergic reactions are IgE-mediated. Complement activation by immune complexes or through the alternative complement pathway has been shown to release complement C3a and C5a anaphylatoxins, which are capable of producing similar reactions.

Autoimmune disease is thought to be caused by antibody or T cell sensitization with autologous self-antigens (see Chapter 28). Postulated mechanisms of this process include the following:

Autoimmune Lymphoproliferative Syndrome

Autoimmune lymphoproliferative syndrome (ALPS) is a disease in which a genetic defect in programmed cell death, or apoptosis, leads to breakdown of lymphocyte homeostasis and normal immunologic tolerance. ALPS is the first pediatric syndrome described in which the primary defect is in apoptosis.

Defective apoptosis in lymphocytes (and, in ALPS type II, dendritic cells) leads to accumulation of these cells in the lymphoid organs after they would normally be eliminated. As a result, cells with autoimmune potential are unchecked and can induce a variety of autoimmune diseases; the risk for malignant transformation to lymphoma is increased.

Patients with ALPS have chronic enlargement of the spleen and lymph nodes, various manifestations of autoimmunity, and elevation of a normally rare population of double-negative T cells (DNTs). When lymphocytes from ALPS patients are cultured in vitro, they are resistant to apoptosis, as compared with cells from healthy controls.

Most ALPS patients have mutations in a TNF receptor gene that is a member of a superfamily (TNFRSF6). This gene, previously known as APT1, encodes the cell surface receptor for the major apoptosis pathway in mature lymphocytes. This receptor has many names, including Fas. The Fas apoptotic pathway is important for eliminating excess T cells after they have been activated and also eliminating antigen-driven and autoreactive T cell clones. Fas is a functional trimer residing at the cell membrane that when engaged by trimeric Fas ligand (FasL), initiates a proteolytic cascade leading to chromosomal DNA degradation and cell death.

CASE STUDY

Laboratory Data

Assay Patient’s Result Reference Range
Total leukocytes 7.2 × 109/L 4.5-9.0 × 109/L
Total lymphocytes 3.2 × 109/L 2.7-5.4 × 109/L
 T lymphocytes 3.15 × 109/L 2.7-5.3 × 109/L
 B lymphocytes Too low to count, almost undetectable
Serum Immunoglobulins
 IgM 0.03 g/L 3.0-15.8 g/L
 IgG 0.31 g/L 0.4-2.2 g/L
 IgA Not detectable 0.15-1.3 g/L
 IgE Not detectable <100 IU/mL
Assay Patient
Blood group O
Anti-A and anti-B titer 1:2, very low
Serum immunoglobulin (mg/dL)  
 IgM 45, low
 IgG 200, very low
 IgA 23, very low

image

image Assessment of Cellular Immune Status

Transplantation Immune Cell Function Assay ImmuKnow, Cylex Inc., Columbia, MD

Principle

Phytohemagglutinin (PHA) is a nonspecific mitogen that can be used to stimulate cell division in CD4 T lymphocytes, regardless of their antigenic specificity or memory status. Therefore, PHA is considered to be a global stimulator of the immune system. The production of intracellular adenosine triphosphate (ATP) is one of the first steps in cellular activation following stimulation with mitogens such as PHA. ATP is a multifunctional nucleotide that plays an indispensable role in the transfer of intracellular chemical energy.

When a sample of patient blood is incubated with PHA, increased ATP production (see figure below) occurs within PHA-activated CD4 T cells. These cells are then isolated by the addition of magnetic beads coated with anti-CD4 monoclonal antibody. The isolated CD4 cells are washed on a magnetic tray and lysed to release intracellular ATP. The amount of measured light emitted following the addition of a luminescence reagent is proportional to the amount of ATP present. An established calibration curve is used to characterize the cellular immune function of the sample.

Results

ATP Level Results Risk of Infection Risk of Rejection
Low Increased Decreased
Moderate Normal Decreased
Strong Normal Increased

Adapted from Associated Regional and University Pathologists (ARUP) Laboratories, 2012 (http://www.aruplab.com).

Chapter Highlights

• Lymphocytes represent the cellular components of the specific system of body defense. These cells function cooperatively in cell-mediated or humoral immunity.

• The primary lymphoid organs in mammals are the bone marrow (and/or fetal liver) and thymus.

• The secondary lymphoid tissues include the lymph nodes, spleen, and Peyer’s patches in the intestine. Proliferation of the T and B lymphocytes in the secondary or peripheral lymphoid tissues is primarily dependent on antigenic stimulation.

• Several major categories of lymphocytes are recognized by the presence of cell surface membrane markers. These categories are T and B cells and natural killer (NK) and K-type lymphocytes.

• The function of plasma cells is the synthesis and excretion of immunoglobulins.

• Monoclonal antibody (MAb) testing led to the present identification of surface membrane markers. Relating MAbs to cell surface antigens now provides a method for classifying and identifying specific cellular membrane characteristics. The current method of testing uses flow cytometry with immunofluorescence.

image

• Immunologic disorders can be divided into primary, secondary (acquired), and those mediated through immune mechanisms. Diseases associated with a primary immunodeficiency are comprised of 40% T cell disorders, 50% B cell disorders, 6% phagocytic abnormalities, and 4% complement alterations. The most common T cell deficiency states are those associated with a concurrent B cell abnormality.