Autoimmunity and Autoimmune Disease

Published on 18/02/2015 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: 3 (2 votes)

This article have been viewed 1486 times

Chapter 20 Autoimmunity and Autoimmune Disease

Summary

Autoimmunity is associated with disease. Autoimmune mechanisms underlie many diseases, some organ-specific, others systemic in distribution, and autoimmune disorders can overlap – an individual may have more than one organ-specific disorder, or more than one systemic disease.

Genetic factors play a role in the development of autoimmune diseases. Twin studies show that there is a heritable component to autoimmunity. The vast majority of autoimmune diseases are polygenic but HLA genes are particularly important.

Self-reactive B and T cells persist even in normal subjects. Autoreactive B and T cells persist in normal subjects, but in disease are selected by autoantigen in the production of autoimmune responses.

Controls on the development of autoimmunity can be bypassed. Microbial cross-reacting antigens and cytokine dysregulation can lead to autoimmunity.

In most diseases associated with autoimmunity, the autoimmune process produces the lesions. The pathogenic role of autoimmunity can be demonstrated in experimental models. Human autoantibodies can be directly pathogenic. Immune complexes are often associated with systemic autoimmune disease. Autoantibody tests are valuable for diagnosis and sometimes for prognosis.

Treatment of autoimmune disease has a variety of aims. Treatment of organ-specific diseases usually involves metabolic control. Treatment of systemic diseases includes the use of anti-inflammatory and immunosuppressive drugs. Biological therapies using monoclonal antibodies against pro-inflammatory cytokines have revolutionized the treatment of the autoimmune rheumatic diseases. B cell-directed therapies have proved highly effective in many autoimmune diseases.

Like a highly trained army, the immune system has evolved to recognize and destroy foreign invading forces. Sometimes immune recognition fails resulting in ‘friendly fire’ against the body’s own tissue. For example, ‘friendly fire’ directed against synovial tissue causes rheumatoid arthritis; whereas an attack on cells within the pancreas results in diabetes mellitus.

Non-specific inflammation (e.g. in response to infection) invariably leads to some degree of ‘collateral’ damage, but because of efficient clearance of the inciting pathogen and negative feedback loops, this is usually self-limited. In contrast, once initiated autoimmune reactions usually persist because the inciting self-antigen cannot be cleared without complete destruction of the target tissue. Furthermore, the tissue destruction resulting from the autoimmune attack may expose previously hidden antigens, leading to further autoantibody production. This phenomenon is known as epitope spread.

Autoimmunity and autoimmune disease

Because the repertoire of specificities expressed by the B cells and T cells is generated randomly, it includes many that are specific for self components. The body therefore requires self-tolerance mechanisms to distinguish between self and non-self determinants to avoid autoreactivity (see Chapter 19). However, such mechanisms may fail and a number of diseases have been identified in which there is autoimmunity, with copious production of autoantibodies and autoreactive T cells. The targets of the autoimmune ‘attacks’ vary widely as we shall see.

Not every ‘autoimmune event’ leads to clinically overt disease. For example, anti-nuclear antibodies may be observed in the healthy relatives of patients with systemic lupus eythematosus, as well as a small number of unrelated, healthy individuals.

Autoimmunity strictly refers to an inappropriate adaptive immune response, i.e. a loss of self-tolerance. Although the terms autoinflammatory and autoimmune are often used interchangeably, the two are not synonymous. Autoinflammatory diseases can occur without autoimmunity. For example, the periodic fever syndromes, which include familial Mediterranean fever and tumor necrosis factor receptor associated periodic syndrome (TRAPS), are caused by dysregulation of the innate immune system, without any adaptive immune response against ‘self’. Conversely, autoimmune diseases need not be inflammatory; examples include immune thrombocytopenia, hemolytic anemia and myasthenia gravis.

Autoimmune conditions present a spectrum between organ-specific and systemic disease

Hashimoto’s thyroiditis is highly organ-specific

One of the earliest examples in which the production of autoantibodies was associated with disease in a given organ is Hashimoto’s thyroiditis. (Thyroiditis is a condition that is most common in middle-aged women and often leads to the formation of a goiter or hypothyroidism.) The gland is infiltrated, sometimes to an extraordinary extent, with inflammatory lymphoid cells. These are predominantly mononuclear phagocytes, lymphocytes and plasma cells, and secondary lymphoid follicles are common (Fig. 20.1). The gland often also has regenerating thyroid follicles.

The serum of patients with Hashimoto’s disease usually contains antibodies to thyroglobulin. These antibodies are demonstrable by agglutination and by precipitin reactions when present in high titer. Most patients also have antibodies directed against a cytoplasmic or microsomal antigen, also present on the apical surface of the follicular epithelial cells (Fig. 20.2), and now known to be thyroid peroxidase, the enzyme that iodinates thyroglobulin. The antibodies associated with Hashimoto’s thyroiditis react only with the thyroid, so the resulting lesion is highly localized.

An individual may have more than one autoimmune disease

Autoimmune diseases ‘hunt in packs’; in patients with an autoimmune disease, the chance of developing an additional autoimmune disease is significantly elevated. This holds true for both organ-specific and systemic autoimmune disease.

The systemic autoimmune rheumatic diseases show considerable overlap. This is typified by the so called ‘overlap syndromes’ in which patients exhibit mixed features of SLE, myositis, and scleroderma. Features of rheumatoid arthritis, notably an erosive arthritis, are present in 5% of patients with SLE. In these diseases immune complexes are deposited systemically, particularly in the kidney, joints, and skin, giving rise to widespread lesions.

The mechanisms of immunopathological damage vary depending on where the disease lies in the spectrum:

Genetic factors in autoimmunity

There is an undoubted familial incidence of autoimmunity. This is largely genetic rather than environmental, as may be seen from studies of identical and non-identical twins. Thus if one of a pair of identical twins gets SLE, there is a 25% chance the other will develop it. If the twin is not identical the concordance rate is only 2–3%.

Within families, clustering of distinct autoimmune diseases has been reported. A large population-based survey found that families with a proband with rheumatoid arthritis were more likely to manifest other autoimmune disorders. This finding holds true for relatives of patients with other autoimmune diseases, such as multiple sclerosis, which suggests the presence of shared pathogenic factors across the autoimmune diseases. However, the majority of individuals with autoimmune disease will not have an affected first degree relative. Thus whilst genetic factors are important in the pathogenesis of autoimmunity, they are usually not sufficient to cause disease without additional environmental influences.

Within the families of patients with organ-specific autoimmunity, not only is there a general predisposition to develop organ-specific antibodies, other genetically controlled factors tend to select the organ that is mainly affected. Thus, although relatives of patients with Hashimoto’s thyroiditis and families of patients with pernicious anemia both have a higher than normal incidence and titer of thyroid autoantibodies, the relatives of patients with pernicious anemia have a far higher frequency of gastric autoantibodies (Fig. 20.5), indicating that there are genetic factors that differentially select the stomach as the target within these families.

Certain HLA haplotypes predispose to autoimmunity

Further evidence for the operation of genetic factors in autoimmune disease comes from their tendency to be associated with particular HLA specificities (Fig. 20.6). For most autoimmune diseases, the MHC region which is located on the short arm of chromosome 6, provides the strongest genetic component to disease susceptibility. Recent high-density genome mapping in multiple autoimmune diseases has demonstrated complex, multilocus effects that span the entire region, with both shared and unique loci across diseases.

In some autoimmune disorders single HLA genes appear to determine disease susceptibility (e.g. HLA-B27 and ankylosing spondylitis). In others, such as rheumatoid arthritis, complex interactions between alleles at multiple genes within the HLA are involved. Rheumatoid arthritis shows no associations with the HLA-A and -B loci haplotypes, but is associated with a nucleotide sequence known as the ‘shared epitope’ (encoding amino acids 70–74 in the DRβ chain) that is common to DR1 and major subtypes of DR4; the MHC association with rheumatoid arthritis is restricted to patients who are positive for antibodies to citrullinated peptides. The nucleotide sequence of the shared epitope is also present in the dnaJ heat-shock proteins of various bacilli and EBV gp110 proteins, suggesting an interesting possibility for the induction of autoimmunity by a microbial cross-reacting epitope (see below). The plot gets deeper, though, with the realization that HLA-DR molecules bearing this sequence can bind to another bacterial heat-shock protein, dnaK, and to the human analog, namely hsp73, which targets selected proteins to lysosomes for antigen processing.

The haplotype B8, DR3 is common in both the organ-specific diseases and systemic autoimmune diseases like SLE and myositis, though Hashimoto’s thyroiditis tends to be associated more with DR5. Interestingly, for type 1 diabetes mellitus, DQ2/8 heterozygotes have a greatly increased risk of developing the disease (see Fig. 20.6).

Genes of the MHC including HLA-A1, B8, and DR3 have been linked to SLE, although over a dozen other genes (including those regulating interferon α) have shown a stronger linkage.

Genes outside the HLA region also confer susceptibility to autoimmunity

Although HLA risk factors tend to dominate, autoimmune disorders are genetically complex and genome-wide searches for mapping the genetic intervals containing genes for predisposition to disease also reveal a plethora of non-HLA genes (Fig. 20.7) affecting:

One of the commonest genes outside of the HLA region to be associated with autoimmune disease is the protein tyrosine phosphatase gene (PTPN22), which is expressed in lymphocytes. The minor allele of PTPN22 (Trp620) is associated with type 1 diabetes, rheumatoid arthritis, autoimmune thyroiditis and SLE. Crohn’s disease, in contrast, is linked to the more common allele (Arg620). The minor allele results in gain of function and results in inhibition of B and T cell activation and homozygous individuals have a profound defect in lymphocyte receptor signaling. The mechanisms by which this leads to autoimmunity are not clear, but possibilities include failure to delete autoreactive T cells in the thymus, impaired regulatory T cell function, and ineffective clearance of pathogens. It is interesting to note that two alleles with contrasting effects on lymphocyte receptor signaling are associated with distinct autoimmune diseases.image

SLE is associated with multiple gene loci

Genome wide-association studies have identified multiple genetic loci associated with SLE (see Fig. 20.7). Null alleles which cause a deficiency of one of the early complement components (C1q, C2, or C4) strongly predispose individuals to a lupus-like disease, although there is a lower incidence compared to ‘classic’ SLE of certain clinical features, notably glomerulonephritis and serological characteristics (ANA and anti-dsDNA antibodies) in these individuals.

The IL2RA locus (which encodes the high-affinity IL-2 receptor) is associated with SLE, type 1 diabetes, multiple sclerosis, Grave’s disease, and ANCA-associated vasculitis.

Serum interferon alpha activity is a heritable risk factor for SLE and there is a strong linkage between single-nucleotide polymorphisms in the genes encoding tyrosine kinase 2 (TYK2) and interferon regulatory factor 5 (IRIF5). TYK2 binds to the type I IFN receptor complex. IRF5 is induces type I IFN gene expression and is downstream of pattern recognition receptor signaling. Microarray studies of peripheral blood mononuclear cells from patients with SLE show a strong type 1 interferon gene expression. The link between SLE and genes controlling interferon suggests the host response to viruses may be important in the pathogenesis of this disorder.

Using a mouse model of lupus, Wakeland and colleagues showed that some linked genes have a cumulative effect leading to autoimmune disease. Thus mice with the sle1 mutation alone show loss of tolerance to chromatin and develop antinuclear antibodies. Mice with sle3 have polyclonal T and B cell activation, with only mild glomerulonephritis. However, mice with both sle1 and sle3 have a full autoimmune response with antibodies to nuclear antigens, splenomegaly and fatal glomerulonephritis.

Autoimmunity and autoimmune disease

Despite the complex selection mechanisms operating to establish self tolerance during lymphocyte development, the body contains large numbers of lymphocytes, which are potentially autoreactive.

Thus, many autoantigens, when injected with adjuvants, make autoantibodies in normal animals, demonstrating the presence of autoreactive B cells, and it is possible to identify a small number of autoreactive B cells (e.g. anti-thyroglobulin) in the normal population.

Autoreactive T cells are also present in normal individuals, as shown by the fact that it is possible to produce autoimmune lines of T cells by stimulation of normal circulating T cells with the appropriate autoantigen (e.g. myelin basic protein [MBP]) and IL-2.

Progression to autoimmune disease occurs in stages

These findings allow us to conceptualize autoimmune disease as a multistep process. The first stage is predisposition of an individual to autoimmunity by his or her genes, and other factors such as female hormones. The second phase is initiated by an event, probably stochastic or perhaps caused by an environmental trigger such as infection or ultraviolet radiation, leading to loss of self-tolerance and autoantibody production. This however is not alone sufficient to cause disease. A further step is required before progression to a third phase involving tissue damage by the autoimmune attack. This autoimmune attack leads to further release of self-antigens, which are not removed in the normal efficient manner (see below), and propagation of the autoimmune response, resulting in the clinical manifestations of disease. The earliest clinical features of systemic autoimmune diseases such as SLE or RA are usually non-specific such as fatigue or constitutional symptoms. This prodrome typically precedes the development of the classic disease phenotype by weeks to months.

During the propagation phase, not only is there an autoimmune response to an increasing number of autoantigens (demonstrated by the sequential development of multiple autoantibodies in patients with SLE), but also to more epitopes within each antigen – a phenomenon termed epitope spread. Epitope spread can be involve multiple epitopes on the same molecule (intramolecular spread), or epitopes on different molecules associated as part of a macromolecular complex (intermolecular spread). The latter provides a mechanism for how antibodies to non-protein self antigens such as DNA and phospholipid can occur.

Autoimmunity results from antigen-driven self-reactive lymphocytes

Given that autoreactive B cells exist, the question remains whether they are stimulated to proliferate and produce autoantibodies by interaction with autoantigens or by some other means, such as non-specific polyclonal activators or idiotypic interactions (see below and Fig. 20.8).

Evidence that B cells are selected by antigen comes from the existence of high-affinity autoantibodies, which arise through somatic mutation, a process that requires both T cells and autoantigen. In addition, patients’ serum usually contains autoantibodies directed to epitope clusters occurring on the same autoantigenic molecule. Apart from the presence of autoantigen itself, it is very difficult to envisage a mechanism that could account for the co-existence of antibody responses to different epitopes on the same molecule. A similar argument applies to the induction, in a single individual, of autoantibodies to organelles (e.g. nucleosomes and spliceosomes, which appear as blebs on the surface of apoptotic cells) or antigens linked within the same organ (e.g. thyroglobulin and thyroid peroxidase).

The most direct evidence for autoimmunity being antigen driven comes from studies of the Obese strain chicken, which spontaneously develops thyroid autoimmunity. If the thyroid gland (the source of antigen) is removed at birth, the chickens mature without developing thyroid autoantibodies (Fig. 20.9). Furthermore, once thyroid autoimmunity has developed, later removal of the thyroid leads to a gross decline of thyroid autoantibodies, usually to undetectable levels.

Comparable experiments have been carried out in the non-obese diabetic (NOD) mouse, which models human autoimmune diabetes – chemical destruction of the β cells leads to decline in pancreatic autoantibodies.

In organ-specific disorders, there is ample evidence for T cells responding to antigens present in the organs under attack. But in non-organ-specific autoimmunity, identification of the antigens recognized by T cells is often inadequate. However, histone-specific T cells are generated in patients with SLE and histone could play a ‘piggyback’ role in the formation of anti-DNA antibodies by substituting for natural antibody in the mechanism outlined in Figure 20.8.

Another possibility is that the T cells do not ‘see’ conventional peptide antigen (possibly true of anti-DNA responses), but instead recognize an antibody’s idiotype (an antigenic determinant on the V region of antibody).

In this view SLE, for example, might sometimes be initiated as an ‘idiotypic disease’, following the model shown in Figure 20.8. In this scheme, autoantibodies are produced normally at low levels by B cells using germline genes. If these then form complexes with the autoantigen, the complexes can be taken up by APCs (including B cells) and components of the complex, including the antibody idiotype, presented to T cells. Idiotype-specific T cells would then provide help to the autoantibody-producing B cells.

Evidence for the induction of anti-DNA and glomerulonephritis by immunization of mice with the idiotype of germline ‘natural’ anti-DNA autoantibody lends credence to this hypothesis.

The ‘waste disposal’ hypothesis of SLE

Antibodies to nuclear components are the serological hallmark of SLE. The question arising from this observation is how nuclear components normally hidden are detected by the immune system as antigen. The answer appears to lie with apoptosis. There is strong evidence that SLE (and possibly other autoimmune diseases) are diseases of failure of clearance of apoptotic cells, i.e. due to decreased macrophage ‘scavenger’ function. When a cell undergoes apoptosis (programmed cell death), blebs of cellular material are formed on the cell surface. Antigens normally buried deep within the cell (and so not detected by the immune system) are exposed on the cell surface. In healthy individuals these apoptotic cells are efficiently cleared. However, in SLE apoptosis is defective; it has been demonstrated that scavenging of apoptotic debris in vitro by macrophages from lupus patients is less efficient than by macrophages from healthy controls. Thus the antigens contained within the apoptotic blebs may trigger an autoimmune response. Apoptotic blebs vary in size; larger blebs contain antigens including Sm, Mi-2, Ro-60, and La, whilst smaller blebs contain fodrin, Jo-1, Ro-52, and ribosomal P. Antibodies against all of these targets can be found in systemic autoimmune diseases such as SLE, Sjögren’s syndrome, and myositis (Fig. 20.10).

A defective complement pathway may also contribute to ineffective clearance of apoptotic cells as C1q binds to cell debris, allowing macrophages with C1q receptors to engulf the apoptotic cells. Complement deficiency in SLE is usually attributed to consumption as a secondary consequence of immune complex formation. However, it is clear that in a very small number of patients complement deficiency is the cause rather than the effect of SLE. In patients with these rare genetic disorders of deficiency of complement components (including C1q, C2 and C4, as discussed previously), there is a hugely increased risk of developing a lupus-like disease. Reduced clearance of immune complexes by the spleen has been demonstrated in a patient with C2 deficiency and SLE. This problem was correctable with transfusions of fresh-frozen plasma containing C2. The C1q knock-out mouse also provides evidence for the role of complement in clearance of apoptotic cells. This mouse develops a lupus-like glomerulonephritis, and renal biopsy reveals multiple apoptotic fragments.

The clustering of the lupus autoantigens in apoptotic blebs provides an elegant explanation for how epitope spread occurs in SLE. Intermolecular epitope spread occurs when molecules in close physical proximity are taken up by an APC. Uptake of an apoptotic cell by an APC will result in presentation of the antigens from the surface of the apoptotic cell. Thus the apoptotic cell acts as a cellular platform or ‘scaffold’ physically linking the multiple autoantigens targeted by the immune response in SLE.image

Defective clearance of apoptotic cells may induce autoimmunity

imageIn mice deletion of some of the molecules involved in the clearance of apoptotic cells leads to autoantibody production and lupus-like disease. However, not all deletions resulting in defective clearance of apoptotic cells result in autoimmunity. Furthermore, immunization of mice with apoptotic cells does not produce autoimmune disease. This may be accounted for by the immunosuppressive capabilities of apoptotic cells, especially through release of anti-inflammatory cytokines from phagocytic cells. SLE-related autoantibodies and glomerulonephritis resembling human lupus nephritis can however be induced in mice by co-immunization with an apoptotic cell binding protein (human beta2 glycoprotein1) and the powerful adjuvant, LPS. Not only did the mice produce high titres of IgG anti-beta2 glycoprotein1 antibodies, but also antibodies to Ro, La, dsDNA, Sm, and nRNP in sequential order, recapitulating human disease. Knockout of CD28 abrogated these effects, indicating that CD28-mediated co-stimulation of T cells is required. This mouse model provides evidence of intermolecular epitope spread, and suggests that long-lived memory T cells responding to a single protein (beta2 glycoprotein1) are capable of providing help to other auto-antigen specific B cells if the protein interacts with a ‘scaffold’ containing the other autoantigens. Of note, immunization with either LPS or beta2 glycoprotein1 alone did not result in lupus autoantibody production.

Induction of autoimmunity

Normally, naive autoreactive T cells recognizing cryptic self epitopes are not switched on because the antigen is presented on ‘non-professional’ APCs such as pancreatic β-islet cells or thyroid epithelial cells which lack co-stimulator molecules, or because it is presented only at low concentrations on ‘professional’ APCs. However the normal mechanisms that maintain self-tolerance may be bypassed.

Molecular mimicry by cross-reactive microbial antigens can stimulate autoreactive lymphocytes

Infection with a microbe bearing antigens that cross-react with the cryptic self epitopes (i.e. have shared epitopes) will load the professional APCs with levels of processed peptides that are sufficient to activate the naive autoreactive T cells. Once primed, these T cells are able to recognize and react with the self epitope on the non-professional APCs because they:

Cross-reactive antigens that share B cell epitopes with self molecules can also break tolerance, but by a different mechanism. Many autoreactive B cells cannot be activated because the CD4+ TH cells they need are unresponsive, either because:

these TH cells are tolerized at lower concentrations of autoantigens than the B cells; or

because they recognize only cryptic epitopes. However, these ‘helpless’ B cells can be stimulated if the cross-reacting antigen bears a ‘foreign’ carrier epitope to which the T cells have not been tolerized (Fig. 20.12). The autoimmune process may persist after clearance of the foreign antigen if the activated B cells now focus the autoantigen on their surface receptors and present it to normally resting autoreactive T cells, which will then proliferate and act as helpers for fresh B cell stimulation.

Molecular mimicry operates in rheumatic fever

An example of a disease in which such molecular mimicry may operate is rheumatic fever, in which autoantibodies to heart valve antigens can be detected. These develop in a small proportion of individuals several weeks after a streptococcal infection of the throat. Carbohydrate antigens on the streptococci cross-react with an antigen on heart valves, so the infection may bypass T cell self tolerance to heart valve antigens. Historically many concepts about autoimmunity arose because the early immunologists often had a background in infectious diseases, and brought with them ideas about cross-reactivity and molecular mimicry extrapolated from rheumatic fever. However, the evidence for molecular mimicry in most chronic autoimmune diseases is lacking or absent. A notable exception is post-infective polyneuropathy.

There is circumstantial evidence for molecular mimicry in anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis (AAV). Antibodies to lysosomal membrane protein-2 (LAMP-2) are a subtype of ANCA found in most patients with pauci-immune focal necrotizing glomerulonephritis (FNGN). The autoantibodies to LAMP-2 commonly recognize an epitope with 100% homology to the bacterial adhesion FimH. Rats injected with FimH develop antibodies to LAMP-2 and pauci-immune FNGN. Furthermore, many humans with pauci-immune FNGN have evidence of recent infection with fimbriated organisms.

Shared B cell epitopes between Yersinia enterolytica and the extracellular domain of the thyroid stimulating hormone (TSH) receptor have been described.

Cytokine dysregulation, inappropriate MHC expression, and failure of suppression may induce autoimmunity

It appears that dysregulation of the cytokine network can also lead to activation of autoreactive T cells, and as noted earlier genes affecting cytokines and their receptors (e.g. IL2RA) are implicated in autoimmune disease by genome-wide association studies.

One experimental demonstration of this is the introduction of a transgene for interferon-γ (IFNγ) into pancreatic β-islet cells. If the transgene for IFNγ is fully expressed in the cells, MHC class II genes are upregulated and autoimmune destruction of the islet cells results. This is not simply a result of a non-specific chaotic IFNγ-induced local inflammatory milieu because normal islets grafted at a separate site are rejected, implying clearly that T cell autoreactivity to the pancreas has been established.

The surface expression of MHC class II in itself is not sufficient to activate the naive autoreactive T cells, but it may be necessary to allow a cell to act as a target for the primed autoreactive TH cells. It was therefore most exciting when cells taken from the glands of patients with Graves’ disease were found to be actively synthesizing class II MHC molecules (Fig. 20.13) and so were able to be recognized by CD4+ T cells.

In this context it is interesting that isolated cells from several animal strains that are susceptible to autoimmunity are also more readily induced by IFNγ to express MHC class II molecules than cells from non-susceptible strains.

The argument that imbalanced cytokine production may also contribute to autoimmunity receives further support from the unexpected finding that tumor necrosis factor (TNF; introduced by means of a TNF transgene) ameliorates the spontaneous SLE-like disease of F1 (NZB × NZW) mice. Furthermore, serological (and less commonly clinical) features of SLE have developed in humans treated with TNF blockade.

Aside from the normal ‘ignorance’ of cryptic self epitopes, other factors that normally restrain potentially autoreactive cells may include:

Deficiencies in any of these factors may increase susceptibility to autoimmunity.

The feedback loop on TH cells and macrophages through the pituitary–adrenal axis is particularly interesting because defects at different stages in the loop turn up in a variety of autoimmune disorders (Fig. 20.14).

For example, patients with rheumatoid arthritis have low circulating corticosteroid levels compared with controls. After surgery, although they produce copious amounts of IL-1 and IL-6, a defect in the hypothalamic paraventricular nucleus prevents the expected increase in adrenocorticotropin (ACTH) and adrenal steroid output.

There is currently intense interest focused on the role of Tregs. Patients with rheumatoid arthritis, for example, reveal a deficiency of Treg function (see below and Fig. 20.w5).

A subset of CD4 regulatory cells present in young healthy mice of the NOD strain, which spontaneously develop IDDM, can prevent the transfer of disease provoked by injection of spleen cells from diabetic animals into NOD mice congenic for the severe combined immunodeficiency trait; this regulatory subset is lost in older mice.

Pre-existing defects in the target organ may increase susceptibility to autoimmunity

We have already noted the sensitivity of target cells to upregulation of MHC class II molecules by IFNγ in animals susceptible to certain autoimmune diseases. Other evidence also favors the view that there may be a pre-existing defect in the target organ.

In the Obese strain chicken model of spontaneous thyroid autoimmunity, not only is there a low threshold of IFNγ induction of MHC class II expression by thymocytes, but when endogenous TSH is suppressed by thyroxine treatment, the uptake of iodine into the thyroid glands is far higher in the Obese strain than in a variety of normal strains. Furthermore, this is not due to any stimulating effect of the autoimmunity because immunosuppressed animals show even higher uptakes of iodine (Fig. 20.15).

Interestingly, the Cornell strain (from which the Obese strain was derived by breeding) shows even higher uptakes of iodine, yet these animals do not develop spontaneous thyroiditis. This could be indicative of a type of abnormal thyroid behavior, which in itself is insufficient to induce autoimmune disease, but does contribute to susceptibility in the Obese strain.

Other situations in which the production of autoantigen is affected are:

The post-translational modification of arginine to citrulline, producing a new autoantigen in rheumatoid arthritis, represents yet another mechanism by which autoimmunity can be evoked.

Autoimmune processes and pathology

Autoimmune processes are often pathogenic. When autoantibodies are found in association with a particular disease there are three possible inferences:

Autoantibodies secondary to a lesion (the second possibility) are sometimes found. For example, cardiac autoantibodies may develop after myocardial infarction.

However, sustained production of autoantibodies rarely follows the release of autoantigens by simple trauma. In most diseases associated with autoimmunity, the evidence supports the first possibility, that the autoimmune process produces the lesions.image

The pathogenic role of autoimmunity can be demonstrated in experimental models

imageThe most direct test of whether autoimmunity is responsible for the lesions of disease is to induce autoimmunity deliberately in an experimental animal and see if this leads to the production of the pathological lesions.

Autoimmunity can be induced in experimental animals by injecting autoantigen (self antigen) together with complete Freund’s adjuvant, and this does indeed produce organ-specific disease in certain organs. For example:

In the case of thyroglobulin-injected animals, not only are thyroid autoantibodies produced, but the gland becomes infiltrated with mononuclear cells and the acinar architecture crumbles, closely resembling the histology of Hashimoto’s thyroiditis.

While these animal models show that experimentally-induced autoimmunity can produce pathological lesions, caution needs to be taken when extrapolating evidence from such models to human disease. It is important to distinguish animal models in which autoimmune disease is artificially induced by large doses of mycobacterial adjuvant, from inbred animal strains which spontaneously develop autoimmune disease (e.g. F1 NZB × NZW mice, discussed below). The former probably have very limited relevance to the pathogenic mechanisms in spontaneous autoimmune disease in humans, not least because they are often of short duration. This may account for the failure of many drugs which showed promise in animal models to translate into effective therapeutics in clinical practice.

Examples of spontaneous autoimmunity

It has proved possible to breed strains of animals that are genetically programmed to develop autoimmune diseases closely resembling their human counterparts.

One well-established example mentioned above is the Obese strain chicken, which parallels human autoimmune thyroid disease in terms of:

So it is of interest that, when the immunological status of Obese strain chickens is altered, quite dramatic effects are seen on the outcome of the disease. For example:

T cells therefore play a variety of pivotal roles as mediators and regulators of this disease.

More directly, a diabetogenic CD4+ T cell clone can induce the chronic leukocytic infiltrate of T cells and macrophages, which damages the insulin-producing β cells of the pancreatic islets of Langerhans in the NOD murine model of type 1 diabetes mellitus (Fig. 20.w2).

The role of the T cells in mediating this attack is further emphasized by the amelioration and prevention of disease by treatment of the mice with a non-depleting anti-CD4 monoclonal antibody, which in the presence of the pancreatic autoantigens, insulin, and glutamic acid decarboxylase (GAD) induces specific T cell anergy.

The dependence of yet another spontaneous model, the F1 hybrid of New Zealand Black and White strains, on the operation of immunological processes is aptly revealed by the suppression of the murine SLE (associated with immune complex glomerulonephritis and anti-DNA autoantibodies), by treatment with anti-CD4 antibody or azathioprine (Fig. 20.w3).

Human autoantibodies can be directly pathogenic

When investigating human autoimmunity directly rather than using animal models, it is of course more difficult to carry out experiments. Nevertheless, there is much evidence to suggest that autoantibodies may be important in pathogenesis, as discussed below.

Autoantibodies can give rise to a wide spectrum of clinical thyroid dysfunction

A number of diseases have been recognized in which autoantibodies to hormone receptors may actually mimic the function of the normal hormone and produce disease. Graves’ disease (thyrotoxicosis) was the first disorder in which such anti-receptor antibodies were clearly recognized.

The phenomenon of neonatal thyrotoxicosis provides us with a natural ‘passive transfer’ study, because the IgG antibodies from the thyrotoxic mother cross the placenta and react directly with the TSH receptor on the neonatal thyroid. Many babies born to thyrotoxic mothers and showing thyroid hyperactivity have been reported, but the problem spontaneously resolves as the antibodies derived from the mother are catabolized in the baby over several weeks.

Whereas autoantibodies to the TSH receptor may stimulate cell division and/or increase the production of thyroid hormones, others can bring about the opposite effect by inhibiting these functions, a phenomenon frequently observed in receptor responses to ligands that act as agonists or antagonists.

Different combinations of the various manifestations of thyroid autoimmune disease – chronic inflammatory cell destruction, and stimulation or inhibition of growth and thyroid hormone synthesis – can give rise to a wide spectrum of clinical thyroid dysfunction (Fig. 20.16).

A variety of other diseases are associated with autoantibodies

Myasthenia gravis provides an example of a disease where some of the autoantibodies can act as a receptor antagonist, blocking the acetyl choline receptor on the post-synaptic membrane of the neuromuscular junction, thus causing muscle weakness and fatigability. A parallel with neonatal hyperthyroidism has been observed – antibodies to acetylcholine receptors from mothers who have myasthenia gravis cross the placenta into the fetus and may cause transient muscle weakness in the newborn baby.

A similar phenomenon is seen in 5% of women who have anti-Ro antibodies (which are found in both SLE and Sjögren’s syndrome). These antibodies can cross the placenta into the fetal circulation causing heart-block and/or transient lupus-like rash in the neonate, providing direct evidence of their pathogenity. This is known as the neonatal lupus syndrome.

ANCA are thought to be directly pathogenic in ANCA-associated vasculitides by causing neutrophil degranulation leading to endothelial damage. ANCA directed against myeloperoxidase (MPO) are commonly found in microscopic polyangiitis. Knockout mice lacking murine MPO (muMPO) can be immunized with muMPO to produce antibodies to muMPO and passive transfer of these antibodies into suitable recipients produces necrotizing arteritis, focal necrotizing crescenteric glomerulonephritis and alveolar capillaritis and hemorrhage reminiscent of human disease.

Somewhat rarely, autoantibodies to insulin receptors and to α-adrenergic receptors can be found, the latter associated with bronchial asthma.

Neuromuscular defects can be elicited in mice injected with serum containing antibodies to presynaptic calcium channels from patients with the Lambert–Eaton syndrome, while sodium channel autoantibodies have been identified in the Guillain–Barré syndrome.

Yet another example of autoimmune disease is seen in rare cases of male infertility where antibodies to spermatozoa lead to clumping of spermatozoa, either by their heads or by their tails, in the semen.

Antibodies to the glomerular capillary basement membrane cause Goodpasture’s disease

Goodpasture’s disease is characterized clinically by rapidly progressive glomerulonephritis and pulmonary haemorrhage. Patients with Goodpasture’s disease have circulating antibodies to the glomerular capillary basement membrane which bind to the kidney and lung (see Fig. 25.3). Evidence for the direct pathogenity of these antibodies was demonstrated by the passive transfer of antibodies eluted from renal biopsy specimens into primates (whose renal antigens were similar to humans). The injected monkeys subsequently died from glomerulonephritis. Subsequent work has shown that these antibodies bind to the several non-collagenous-1 (NC-1) domains of type IV collagen in the GBM. Moreover, immunization of animals with NC-1 domains induces glomerulonephritis, providing a causal link between autoantigen and antibody.

Immune complexes appear to be pathogenic in systemic autoimmunity

In the case of SLE, it can be shown that complement-fixing complexes of antibody with DNA and other nucleosome components such as histones are deposited in the kidney (see Fig. 25.3), skin, joints, and choroid plexus of patients, and must be presumed to produce type III hypersensitivity reactions as outlined in Chapter 25. A variety of different antibodies have been eluted from the kidney biopsies of patients with SLE. These include anti-dsDNA (nucleosomes), anti-Ro and anti-Sm/RNP. Whilst placing these antibodies at the scene of the crime, their mere presence does not prove they ‘pulled the trigger’. However, experiments using murine monoclonal anti-dsDNA antibodies in a rat kidney perfusion system, and other evidence from the use of human hybridoma derived anti-dsDNA antibodies in SCID mice, provides compelling evidence that some anti-dsDNA antibodies are genuinely pathogenic.

It has been proposed that anti-dsDNA/nucleosome antibodies bind to the negatively charged surface of the renal glomerulus via a histone (positively charged) ‘bridge’. The histone is part of the nucleosome complex. The formation of immune complexes at the glomerular surface membrane is thought to induce an inflammatory response leading to the kidney damage so frequently seen in patients with SLE, though the precise mechanisms have yet to be elucidated.

Autoantibodies to IgG provoke pathological damage in rheumatoid arthritis

The erosions of cartilage and bone in rheumatoid arthritis are mediated by macrophages and fibroblasts, which become stimulated by cytokines from activated T cells and immune complexes generated by a vigorous immunological reaction within the synovial tissue (Fig. 20.19).

The complexes can arise through the self-association of IgG rheumatoid factors specific for the Fcγ domains – a process facilitated by the striking deficiency of terminal galactose on the biantennary N-linked Fc oligosaccharides (Fig. 20.20). This agalacto glycoform of IgG in complexes can exacerbate inflammation through reaction with mannose-binding lectin and production of TNFα.

Evidence for directly pathogenic T cells in human autoimmune disease is hard to obtain

Adoptive transfer studies have shown that TH1 cells are responsible for directly initiating the lesions in experimental models of organ-specific autoimmunity.

In humans, evidence for a pivotal role of T cells in the development of autoimmune disease includes:

However, it is difficult to identify a role for the T cell as a pathogenic agent as distinct from a TH function in the organ-specific disorders.

Indirect evidence from circumstances showing that antibodies themselves do not cause disease, such as in babies born to mothers with type 1 diabetes mellitus, may be indicative.

TH17 cells – a new player in autoimmunity?

Recent interest has focused on the role of TH17 cells in auto-immunity. Their initial discovery came about when it was noted that deletion of key TH1 molecules such as IL12 and IFN-γR did not abrogate EAE and collagen induced arthritis (CIA) in mice. In fact, these animals showed enhanced susceptibility, casting doubt on the role of TH1 cells as the fundamental players in autoimmunity. This led to the identification of a new cytokine IL23 – knockout of IL23 is protective against experimentally induced autoimmunity and IL23 was subsequently shown to induce IL-17 from activated T cells. Such IL-23 driven TH cells show a unique pattern of gene expression (which differs from that of IL-12 driven TH1 cells), and are now known to be TH17 cells. Whilst TGFβ, IL1β, and IL6, and not IL23, are the key cytokines for the induction of TH17 cells, IL23 is important for their maintenance; IL-23 deficient mice show normal numbers of TH1 cells but a reduction in TH17 cells.

In humans, high levels of IL-17 and its receptor are found in the synovial fluid and tissue of patients with RA. However, the number of TH17 cells is not elevated in the synovial fluid or PBMCs with RA compared to healthy controls. Multiple SNPs in the IL23 receptor gene region, as well as other genes involved in the IL23/TH17 pathway, are associated with inflammatory bowel disease. The results of trials of anti-IL-17 monoclonal antibodies in patients with RA and MS will help clarify the role of TH17 cells in these diseases.

Autoantibodies for diagnosis, prognosis, and monitoring

Whatever the relationship of autoantibodies to the disease process, they frequently provide valuable markers for diagnostic purposes. A particularly good example is the test for mitochondrial antibodies, used in diagnosing primary biliary cirrhosis (Fig. 20.21). Historically, exploratory laparotomy was needed to obtain this diagnosis, and was often hazardous because of the age and condition of the patients concerned. Some other diagnostically useful antibodies are listed below:

Prognosis and disease subtype:

Disease monitoring:

Treatment of autoimmune diseases

Many autoimmune diseases can be treated successfully. Often, in organ-specific autoimmune disorders, the symptoms can be corrected by metabolic control. For example:

If the target organ is not completely destroyed, it may be possible to protect the surviving cells by transfection with FasL or TGFβ genes.

Where function is completely lost and cannot be substituted by hormones, as may occur in lupus nephritis or chronic rheumatoid arthritis, tissue grafts or mechanical substitutes may be appropriate. In the case of tissue grafts, protection from the immunological processes that necessitated the transplant may be required.

Conventional immunosuppressive therapy with anti-mitotic drugs at high doses can be used to damp down the immune response, but, because of the dangers involved, tends to be used only in organ or life-threatening disorders such as SLE, myositis and ANCA-associated vasculitis (AAV).

Advances in treatment have transformed the 5 year survival rate in severe systemic autoimmune diseases such as SLE and AAV from around 50% and less than 10% respectively in the mid 20th century to over 90% today. However, the costs of this success in controlling autoimmune disease activity are the adverse effects of immunosuppressants, especially glucocorticoids. Most of the early mortality in SLE or AAV is now due to infection secondary to immunosuppressive therapy rather than from uncontrolled autoimmune disease. Long term use of drugs such as cyclophosphamide and azathioprine also increases the risk of infertility and possibly malignancy. The challenge now is to minimize treatment toxicity. This is likely to be achieved through several means. Firstly, the rational design of targeted therapies (in contrast to the ‘shot-gun’ approach of non-specific cytotoxics such as cyclophosphamide) should reduce the toxicity of treatment regimes. Secondly, it should become possible to identify patients through means of biomarkers in whom immunosuppression can be safely reduced. Some such biomarkers have already been identified but require further validation before translation into clinical practice. For example, CD8+ T cell transcription signatures from blood samples taken at time of diagnosis in patients with AAV and SLE identify subgroups of patients with high and low risk of subsequent relapse.

The most recent treatments for autoimmune disease are antibodies that target individual elements of the immune system, including:

‘Biologics’ – a new paradigm in the treatment of autoimmune rheumatic disease

imageThe treatment of patients with rheumatoid arthritis has been revolutionized by the introduction of biological agents. Infliximab, a murine-human chimeric antibody against TNFα, not only markedly alleviates the symptoms of RA such as joint pain, stiffness and swelling, but also halts the progression of joint destruction (Fig. 20.w4). A number of other anti-TNF agents have subsequently become available including adalimumab (a fully humanized monoclonal antibody against TNF), etanercept (a soluble TNF receptor), and certolizumab (a pegylated anti-TNF antibody). It is fascinating to record that the compromised regulatory T cell function in the patients is reversed by such therapy (Fig. 20.w5). Similarly impressive results in RA are seen with B-cell depletion using rituximab, an anti-CD20 monoclonal antibody, and tocilizumab, a monoclonal antibody against IL-6. Other biologics licensed for use in RA include abatacept, which inhibits the co-stimulatory interaction between T cells and APCs, and anakinra, an IL-1 blocker.

Rituximab has shown notable efficacy across the spectrum of autoimmune disease from ITP to multiple sclerosis. Randomized controlled trials show that it has equivalent efficacy to cyclophosphamide in AAV. It is particularly useful in patients with AAV who are refractory to cyclophosphamide treatment. Rituximab treatment results in reduction of the titer of the pathogenic anti-neutrophil cytoplasmic antibodies. Despite initial encouraging results from uncontrolled series, two randomized controlled trials did not demonstrate benefit from rituximab in SLE. However, this may be accounted for by poor trial design. Belimumab, which targets the B-cell survival factor BAFF (also known as BLyS), has, in contrast to rituximab, demonstrated efficacy in SLE in two randomized trials. In the future, the combination of an anti-CD20 agent such as rituximab with anti-BLys therapy using belimumab may produce longer lasting disease remission.

Less well-established approaches to treatment may become practicable

As we understand more about the molecular mechanisms underlying autoimmunity, targeted therapy is becoming increasingly possible (Fig. 20.22):

several centers are trying out autologous stem cell transplantation following hematoimmunoablation with cytotoxic drugs for patients with severe SLE and vasculitis;

repeated injection of Cop 1 (a random copolymer of alanine, glutamic acid, lysine, and tyrosine) reduces relapse rate in relapsing–remitting multiple sclerosis. Cop 1 was originally designed to simulate the postulated ‘guilty’ autoantigen, MBP, and induce experimental autoimmune encephalitis; paradoxically it had the opposite effect. This suggests it is possible to achieve antigen-specific immune suppression.

eculizumab, a monoclonal antibody against complement component C5, has been used successfully in paroxysmal nocturnal hemoglobinuria. It may prove effective in SLE;

TH cell subsets show considerable plasticity in vivo. Manipulation of the balance between pro-inflammatory TH17 cells and anti-inflammatory regulatory T cells may prove fruitful. Therapeutic interventions to increase the numbers of Tregs might increase self-tolerance and ‘put the brakes’ on inflammatory cascades. Adoptive transfer of polyclonal Tregs can prevent the onset of diabetes in NOD mice. However, it has become clear that successful treatment of established autoimmune disease by adoptive transfer of Tregs is antigen-specific; only Tregs specific for pancreatic antigens (and not polyclonal T regs) can reverse diabetes in NOD mice. This presents difficulties for treatment in human disease, where the inciting antigen is rarely known;

inhibition of the activation or differentiation of TH17 cells, and/or blockade of linked cytokines such as IL17A and IL-23 may prove valuable. Trials of anti-IL17 monoclonal antibodies are already underway;

small molecule therapies have the advantage of being cheaper to manufacture than monoclonal antibodies, of particular relevance in an era of spiraling healthcare costs. Kinases are intracellular enzymes that play a crucial role in signal transduction pathways controlling a number of cellular functions. Kinase inhibitors have been successfully used in oncology, but may also prove effective in autoimmune diseases. For example, a splenic tyrosine kinase inhibitor, reduces severity of antibody-mediated glomerulonephritis in rats, and is being trialled in RA.

The prospects for these new treatments either individually or in combination with therapeutic antibodies is very positive.

Critical thinking: Autoimmunity and autoimmune disease (see p. 440 for explanations)

Miss Jacob, a 30-year-old Caribbean woman, was seen in a rheumatology clinic with stiff painful joints in her hands, which were worse first thing in the morning. Other symptoms included fatigue, a low-grade fever, a weight loss of 2 kg, and some mild chest pain. Miss Jacob had recently returned to the UK from a holiday in Jamaica and was also noted to be taking the combined oral contraceptive pill. Past medical history of note was a mild autoimmune hemolytic anemia 2 years previously.

On examination Miss Jacob had a non-specific maculopapular rash on her face and chest, and patchy alopecia (hair loss) over her scalp. Her mouth was tender and examination revealed an ulcer on the soft palate. She had moderately swollen and tender proximal interphalangeal joints. Her other joints were unaffected, but she had generalized muscle aches. The results of investigations are shown in the table.

Investigation Result
radiograph of hands soft tissue swelling, but no bone erosions
chest radiograph a small pleural effusion at the right lung base
full blood count a mild normocytic, normochromic anemia and mild lymphocytopenia
C-reactive protein levels normal
erythrocyte sedimentation rate raised
rheumatoid factor negative
serum IgG levels raised
anti-nuclear antibodies (ANA) positive by immunofluorescence
anti-double-stranded DNA, anti-RNA, and anti-histone positive by ELISA antibodies
complement (C3 and C4) levels low
skin biopsy from an area unaffected by the rash deposition of IgG and complement components at the junction between dermis and epidermis (lupus ‘band’ test)

A diagnosis of systemic lupus erythematosus (SLE) was made. Miss Jacob was treated with hydroxychloroquine, an anti-malarial, for the rash and the arthritis.

At a follow-up appointment urinalysis showed protein and red cells. Serum creatinine was mildly elevated as was her blood pressure. A renal biopsy showed membranous lupus nephritis. She was prescribed oral corticosteroids, mycophenolate mofetil and an angiotensin converting enzyme (ACE) inhibitor, which improved her renal function and blood pressure. Her physician also gave advice regarding birth control and pregnancy, and regular check-ups were arranged.

Further reading

Arbuckle M.R., McClain M.T., Rubertone M.V., et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003;349:1526–1533.

Betterle C., Greggio N.A., Volpato M. Clinical Review 93: Autoimmune polyglandular syndrome type 1. J Clin Endocrinol Metab. 1998;83:1049–1055.

Edwards J.C., Szczepanski L., Szechinski J., et al. Efficacy of the novel B cell targeted therapy, rituximab, in patients with active rheumatoid arthritis. N Engl J Med. 2004;350:2572–2581.

Damsker J.M., Hansen A.M., Caspi R.R. TH1 and TH17 cells: adversaries and collaborators. Ann N Y Acad Sci. 2010;1183:211–221.

Rioux J.D., Goyette P., Vyse T.J., et alInternational MHC and Autoimmunity Genetics Network. Mapping of multiple susceptibility variants within the MHC region for 7 immune-mediated diseases. Proc Natl Acad Sci USA. 2009;106:18680–18685.

Keymeulen B., Vandemeulebroucke E., Ziegler A.G., et al. Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J Med. 2005;352:2598–2608.

Levine J.S., Subang R., Nasr S.H., et al. Immunization with an apoptotic cell-binding protein recapitulates the nephritis and sequential autoantibody emergence of systemic lupus erythematosus. J Immunol. 2006;177:6504–6516.

McGaha T.L., Sorrentino B., Ravetch J.V. Restoration of tolerance in lupus targeted inhibiting receptor expression. Science. 2005;307:590–593.

Notley C.A., Ehrenstein M.R. The yin and yang of regulatory T cells and inflammation in RA. Nat Rev Rheumatol. 2010;6:572.

Park H., Li Z., Yang X.O., et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005;6:1133–1141.

Rahman A., Isenberg D.A. Systemic lupus erythematosus. N Engl J Med. 2008;358:929–939.

Roitt I.M., Doniach D., Campbell P.N., Vaughan-Hudson R. Auto-antibodies in Hashimoto’s disease (lymphadenoid goitre). Lancet. 1956(ii):820–821.

Suber T., Rosen A. Apoptotic cell blebs: repositories of autoantigens and contributors to immune context. Arthritis Rheum. 2009;60:2216–2219.

Wakeland E.K., Liu K., Graham R.R., Behrens T.W. Delineating the genetic basis of systemic lupus erythematosus. Immunity. 2001;15:397–408.

Zenewicz L.A., Abraham C., Flavell R.A., Cho J.H. Unraveling the genetics of autoimmunity. Cell. 2010;140:791–797.

Further references

Botto M., Dell’Agnola C., Bygrave A.E., et al. Homozygous C1q deficiency causes glomerulonephritis associated with multiple apoptotic bodies. Nat Genet. 1998;19:56–59.

Carr E.J., Clatworthy M.R., Lowe C.E., et al. Contrasting genetic association of IL2RA with SLE and ANCA-associated vasculitis. BMC Med Genet. 2009;10:22.

Davies K.A., Erlendsson K., Beynon H.L., et al. Splenic uptake of immune complexes in man is complement dependent. J Immunol. 1993;151:3866–3873.

Ding B., Padyukov L., Lundström E., et al. Different patterns of associations with anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis in the extended major histocompatibility complex region. Arthritis Rheum. 2009;60:30–38.

Herrmann M., Voll R.E., Zoller O.M., et al. Impaired phagocytosis of apoptotic cell material by monocyte-derived macrophages from patients with systemic lupus erythematosus. Arthritis Rheum. 1998;41:1241–1250.

Isenberg D.A., Manson J.J., Ehrenstein M.R., et al. Anti-dsDNA antibodies- at journey’s end? Rheumatology. 2007;46:1052–1056.

Kain R., Exner M., Brandes R., et al. Molecular mimicry in pauci-immune focal necrotizing glomerulonephritis. Nat Med. 2008;14:1088–1096.

Lerner R.A., Glassock R.J., Dixon F.J. The role of anti-glomerular basement membrane antibody in the pathogenesis of human glomerulonephritis. J Exp Med. 1967;126:989–1004.

Lin J.P., Cash J.M., Doyle S.Z., et al. Familial clustering of rheumatoid arthritis with other autoimmune diseases. Hum Genet. 1998;103:475–482.

Mattey D.L., Dawes P.T., Clarke S., et al. Relationship among the HLA-DRB1 shared epitope, smoking, and rheumatoid factor production in rheumatoid arthritis. Arthritis Rheum. 2002;47:403–407.

McKinney E., Lyons P.A., Carr E.J., et al. A CD8+ T cell transcription signature predicts prognosis in autoimmune disease. Nat Med. 2010;16:586–591.

Niewold T.B., Hua J., Lehman T.J., et al. High serum IFN-alpha activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun. 2007;8:492–502.

Pinching A.J., Rees A.J., Pussell B.A., et al. Relapses in Wegener’s granulomatosis: the role of infection. Br Med J. 1980;281:836–838.

Rioux J.D., Goyette P., Vyse T.J., et al. Mapping of multiple susceptibility variants within the MHC region for 7 immune-mediated diseases. Proc Natl Acad Sci USA. 2009;106:18680–18685.

Silman A.J., Newman J., MacGregor A.J. Cigarette smoking increases the risk of rheumatoid arthritis: Results from a nationwide study of disease-discordant twins. Arthritis Rheum. 1996;39:732–735.

Stone J.H., Merkel P.A., Spiera R., Seo P., et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. 2010;363:221–232.

Tan E.M., Feltkamp T.E., Smolen J.S., et al. Range of antinuclear antibodies in “healthy” individuals. Arthritis Rheum. 1997;40:1601–1611.

Wakeland E.K., Liu K., Graham R.R., Behrens T.W. Delineating the genetic basis of systemic lupus erythematosus. Immunity. 2001;15:397–408.

Weinblatt M., Kavanaugh A., Genovese M., et al. An oral spleen tyrosine kinase (Syk) inhibitor for rheumatoid arthritis. N Engl J Med. 2010;363:1303–1312.

Yamada H., Nakashima Y., Okazaki K., et al. TH1 but not TH17 cells predominate in the joints of patients with rheumatoid arthritis. Ann Rheum Dis. 2008;67:1299–1304.