Fact-checked by Grok 2 weeks ago

Autoimmunity

Autoimmunity is a pathological condition in which the fails to distinguish self from non-self antigens, resulting in the production of autoantibodies and autoreactive T cells that attack the body's own healthy cells and tissues, often leading to chronic and tissue damage. This aberrant underlies a diverse group of disorders known as autoimmune diseases, which can manifest systemically across multiple organs or be confined to specific tissues. Autoimmune diseases represent a significant burden, affecting an estimated 8% of the U.S. population, or approximately 24 to 50 million individuals, with women disproportionately impacted—comprising about 78% of cases due to potential hormonal and genetic factors. More than 80 distinct autoimmune disorders have been identified, ranging from common conditions like and systemic lupus erythematosus to organ-specific diseases such as mellitus and . These diseases often develop insidiously over time, with symptoms varying widely based on the affected tissues, including joints, skin, endocrine glands, and blood vessels, and they frequently coexist in individuals with genetic predispositions. The of autoimmunity is multifactorial, involving a complex interplay of genetic susceptibility—such as specific (HLA) alleles—environmental triggers like infections, toxins, drugs, and light exposure, and hormonal influences that may explain the female predominance. Pathophysiologically, autoimmunity arises from breakdowns in mechanisms, including central tolerance in the and , and in lymphoid organs, allowing autoreactive lymphocytes to escape regulation and initiate self-directed immune attacks. While the precise triggers remain under investigation, recent studies indicate a rising global prevalence, potentially linked to lifestyle changes, pollution, and microbiome alterations, underscoring the need for ongoing into prevention and targeted therapies.

Introduction and Fundamentals

Definition and Overview

Autoimmunity is a pathological condition in which the fails to distinguish self-antigens from foreign ones, resulting in an aberrant against the body's own tissues and cells. This failure leads to the of autoantibodies—such as IgG and IgM directed against self-components—or the of autoreactive T lymphocytes, which mediate and to host structures. Unlike the , which provides rapid, non-specific defense against pathogens through mechanisms like and complement , the relies on antigen-specific recognition by B and T cells to generate targeted responses; in autoimmunity, this specificity is misdirected toward self, often due to a breakdown in immunological . Core to autoimmunity are autoreactive lymphocytes, including B cells that secrete autoantibodies and T cells that orchestrate cellular attacks, which can form immune complexes that deposit in tissues and perpetuate damage. Autoantibodies, particularly IgG isotypes, are often pathogenic by binding to self-antigens and activating complement or recruiting inflammatory cells, while IgM autoantibodies may play roles in both protective and harmful contexts depending on the disease stage. Globally, autoimmune diseases affect approximately 5-10% of the population, manifesting in over 80 distinct conditions that disproportionately impact women and vary by ethnicity and geography. From an evolutionary standpoint, low-level autoimmunity may represent a byproduct of the immune system's for robust against diverse pathogens, where heightened reactivity to foreign threats inadvertently increases the risk of self-reactivity. This trade-off ensures survival advantages in pathogen-rich environments but predisposes modern populations to autoimmune disorders when regulatory mechanisms falter. The consequences of autoimmunity include chronic inflammation, which drives ongoing tissue injury, and progressive organ destruction, as seen in the thyroid gland during autoimmune thyroiditis or in synovial joints affected by . These processes can lead to functional impairments, such as or joint deformities, underscoring the need for mechanisms like central and to prevent such erroneous responses.

Low-Level Autoimmunity

Low-level autoimmunity refers to the physiological presence of transient, low-affinity autoreactive antibodies and T cells that play an essential role in maintaining immune without causing tissue damage. These natural autoantibodies, primarily of the IgM class and encoded by genes, exhibit polyreactivity and moderate affinity for self-antigens, enabling them to bind altered or damaged cellular components. A key example is their involvement in the clearance of apoptotic cells, where they opsonize debris to facilitate and prevent the release of intracellular contents that could trigger . These autoreactive elements contribute to several beneficial functions, including tissue repair, the removal of senescent or damaged cells, and immune surveillance against potential threats. In tissue repair, natural autoantibodies promote regeneration by recognizing oxidation-specific epitopes on injured cells, aiding in the resolution of and . For immune surveillance, they act as a system, binding both self-molecules and microbial patterns to bridge innate and adaptive immunity. Notably, anti-nuclear antibodies (), often present at low titers in healthy individuals with a of up to 20-30%, exemplify this non-pathogenic autoreactivity, as they rarely lead to clinical manifestations in the absence of other factors.31313-0/fulltext) Evidence from comparative studies highlights the basal nature of this autoreactivity. In conventional mice, natural autoantibodies against self-antigens like phosphorylcholine are readily detectable, whereas germ-free mice show significantly reduced levels, underscoring the influence of environmental in shaping physiological autoreactivity without inducing . These low-level responses remain contained and do not progress to pathology in healthy hosts, as demonstrated by the absence of autoimmune symptoms in animals with intact regulatory mechanisms despite detectable autoreactive clones. The distinction between physiological and pathological autoimmunity can be understood through a , where low-affinity, low-level autoreactivity maintains immune balance and , while escalation beyond a critical —due to genetic, environmental, or factors—leads to high-affinity, persistent responses and disease. This model posits that autoreactive cells operate within a tolerable range in healthy states, supported by regulatory checkpoints like T regulatory cells, preventing unchecked expansion. Seminal work on affinity has shown that affinities below this pose minimal risk of autoimmunity, reinforcing the adaptive value of controlled basal autoreactivity.

Historical Development

Early Observations

Ancient medical texts contain some of the earliest descriptions of conditions now recognized as autoimmune diseases, though without understanding of their immune-mediated nature. Around 400 BCE, documented , referring to it as "leuce" or white skin, characterized by depigmented patches (now known to result from autoimmune destruction of melanocytes). Similarly, goiter—an enlargement of the gland—was observed by and later by in the 1st century CE, often attributed to environmental factors like consumption of snowmelt water, but representing early clinical recognition of thyroid pathology later linked to autoimmunity. In the late 19th and early 20th centuries, clinical observations began hinting at immune self-attack mechanisms. In 1900, , in his Croonian Lecture to the Royal Society, coined the term "horror autotoxicus" to describe the immune system's inherent prohibition against producing antibodies that harm the body's own tissues, warning of the potential for self-poisoning if this barrier failed. This concept underscored the prevailing view that autoimmunity was theoretically impossible, yet emerging cases challenged it. For instance, the 1906 for , developed by August von Wassermann and colleagues, detected serum antibodies (reagins) that reacted with beef heart extract containing —a self-lipid—demonstrating cross-reactive autoantibodies in an infectious context. Early 20th-century case reports further illustrated immune-mediated self-damage. Acquired hemolytic anemia, involving destruction of red blood cells, had been noted in clinical descriptions since the late , with cases like those reported by Georges Hayem in 1878 suggesting non-inherited forms of potentially due to toxic or immune factors. By the 1910s, thyroid pathology provided clearer examples; in 1912, described "struma lymphomatosa," a chronic lymphocytic infiltration of the leading to and , based on four surgical cases—findings later confirmed as autoimmune . A pivotal observation came in 1904, when Julius Donath and identified a biphasic in paroxysmal cold (PCH), a rare hemolytic disorder. Their thermal amplitude test showed that patient serum, when incubated with normal red cells at cold temperatures followed by warming, caused complement-mediated —marking the first demonstrated human directly causing disease. These pre-1930s clinical insights, though not framed in modern immunological terms, laid the groundwork for recognizing autoimmunity through patterns of unexplained self-tissue damage.

Key Milestones and Discoveries

In the 1940s, researchers developed experimental models of in rabbits by immunizing them with autologous red blood cells, revealing immune-mediated and the role of the in disease progression. This work laid foundational insights into antibody-dependent destruction of self-cells. In 1948, Harry M. Rose and colleagues identified through differential agglutination tests using sera from patients, demonstrating autoantibodies that bind the Fc region of IgG and advancing recognition of humoral autoimmunity in joint disease. The 1950s brought experimental validation of autoimmunity as a mechanism. In 1956, Noel R. Rose and Ernest Witebsky induced in rabbits and guinea pigs by immunizing them with extracts, overturning Ehrlich's "horror autotoxicus" and providing the first proof that autoimmunity could be deliberately provoked in healthy animals. This breakthrough established organ-specific autoimmunity models. In 1957, George Friou developed the immunofluorescent test for antinuclear antibodies (), facilitating the detection of autoantibodies in systemic lupus erythematosus (SLE) and other autoimmune conditions. Complementing this, Frank Macfarlane Burnet's 1959 proposed that lymphocytes are pre-committed to specific antigens during development, with self-reactive clones eliminated to maintain ; failures in this process could thus trigger autoimmunity. During the 1970s, genetic links to autoimmunity emerged through identification of (HLA) associations with diseases such as (HLA-B27) and (HLA-DR4), underscoring MHC molecules' role in presenting self-antigens to T cells. These findings culminated in the 1980 Nobel Prize in or awarded to George D. Snell, Jean Dausset, and for discoveries concerning MHC structure and function, which elucidated how genetic variations in MHC genes influence immune responses and susceptibility to autoimmune disorders. From the 1980s onward, diagnostic advancements included the 1982 American College of Rheumatology criteria for systemic lupus erythematosus, which incorporated autoantibody panels such as anti-dsDNA and anti-Sm testing to improve classification accuracy. More recently, studies up to 2025 have highlighted the gut microbiome's influence on autoimmunity, particularly in non-obese diabetic (NOD) mice models of type 1 diabetes, where specific bacteria like Akkermansia muciniphila remodel the microbiota to suppress islet autoimmunity via the gut-pancreas axis.

Mechanisms of Immune Regulation

Immunological Tolerance

Immunological tolerance refers to the array of mechanisms that enable the to distinguish self from non-self antigens, thereby preventing autoimmune responses while allowing effective defense against pathogens. These processes are essential for maintaining and occur primarily through central and peripheral pathways. Central tolerance eliminates self-reactive lymphocytes during their development in primary lymphoid organs, while acts on mature lymphocytes that escape central mechanisms, ensuring that low-level autoimmunity remains regulated without causing harm. Central tolerance for T cells takes place in the , where developing thymocytes undergo negative selection to delete those with high-affinity recognition of self-antigens presented by (MHC) molecules. This process involves of self-reactive thymocytes in the thymic medulla, triggered by strong TCR signaling upon encounter with self-peptides on antigen-presenting cells such as dendritic cells and medullary thymic epithelial cells. For B cells, central tolerance occurs in the , where immature B cells expressing self-reactive B cell receptors (BCRs) are subjected to or receptor editing to alter their antigen specificity and avoid autoreactivity. These mechanisms collectively purge the majority of potentially autoreactive clones before they enter circulation. Peripheral tolerance mechanisms complement central tolerance by inactivating or suppressing any remaining self-reactive lymphocytes in secondary lymphoid organs and tissues. A key process is T cell anergy, a state of functional unresponsiveness induced when self-antigens engage the (TCR) without adequate , leading to inhibited proliferation and cytokine production. Regulatory T cells (Tregs), characterized by expression of the transcription factor , play a central role in active suppression by interacting with effector T cells and dendritic cells to dampen inflammatory responses. Their discovery and elucidation of their role in peripheral tolerance were recognized by the 2025 in Physiology or Medicine, awarded to Mary E. Brunkow, Fred Ramsdell, and Shimon Sakaguchi. Tregs exert their effects through cell-contact-dependent mechanisms and of immunosuppressive cytokines such as interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), which inhibit activation and promote tissue repair. Additional peripheral checkpoints include requirements for , where T cell activation demands engagement of on T cells with B7 molecules (/) on antigen-presenting cells; absence of this signal promotes anergy or . The Fas-Fas ligand (FasL) pathway further enforces by inducing in activated self-reactive T cells, particularly during repeated stimulation, preventing their accumulation and potential autoaggression. Breakdown of these tolerance mechanisms can lead to autoimmunity, as exemplified by defects in the (AIRE) gene, which is crucial for promiscuous expression of tissue-specific self-antigens in the thymic medulla to facilitate negative selection. Mutations in AIRE impair this presentation, resulting in the autoimmune polyendocrinopathy-candidiasis-ectodermal (APECED) syndrome, characterized by multi-organ autoimmunity due to failure of central T cell .

Role of Immunodeficiency in Autoimmunity

Autoimmunity paradoxically arises in states of immunodeficiency, where defects in immune regulation fail to prevent self-reactive responses despite overall immune compromise. Primary immunodeficiencies, such as common variable immunodeficiency (CVID), exhibit autoimmunity in 20-30% of cases, often due to impaired regulatory T cells (Tregs) that normally maintain immunological tolerance. This high prevalence highlights how the loss of suppressor mechanisms can tip the balance toward autoreactivity, even as antibody production or other immune functions are diminished. Key mechanisms include the depletion or dysfunction of suppressor cells, such as Tregs, which disrupts central and to self-antigens. Additionally, chronic infections prevalent in immunodeficient states can drive autoimmunity through molecular mimicry, where persistent microbial antigens resemble self-proteins, eliciting cross-reactive immune responses. In primary immunodeficiencies like Wiskott-Aldrich syndrome, this manifests as autoimmune cytopenias, including and , affecting up to 72% of patients through combined defects in T- and B-cell regulation. Similarly, selective IgA deficiency is associated with an increased risk of celiac disease, where mucosal immune dysregulation overlaps with gluten-induced autoimmunity. Secondary immunodeficiencies further illustrate this link, as acquired immune suppression can unmask or provoke autoimmune phenomena. In , progressive T-cell depletion leads to SLE-like syndromes characterized by autoantibodies, cytopenias, and inflammatory features, mimicking systemic due to dysregulated B-cell hyperactivity. Post-chemotherapy states, involving transient secondary from myelosuppression, have also been linked to emergent autoimmunity, such as or , as immune reconstitution favors autoreactive clones amid antigen release from damaged tissues.

Etiology and Risk Factors

Autoimmune diseases exhibit a substantial genetic component, as evidenced by twin studies demonstrating higher concordance rates in monozygotic (MZ) twins compared to dizygotic twins. For (RA), MZ twin concordance rates are approximately 15%, while for (MS), they range from 20% to 25%. These patterns indicate estimates of 50-60% for RA and around 50% for MS, underscoring the role of genetic factors in susceptibility without full . Among the most prominent genetic risk factors are variants in the (HLA) genes, which encode molecules critical for . For instance, alleles, particularly those carrying the shared , confer increased risk for RA with odds ratios of 3 to 5. Similarly, is strongly associated with , present in 90% of affected individuals but only 5-8% of the general population. Non-HLA genes also contribute significantly; PTPN22 encodes a phosphatase that inhibits T-cell activation, and its risk variant (rs2476601) is linked to multiple autoimmune conditions including RA and . CTLA4, which regulates T-cell co-stimulation by acting as a negative regulator, has polymorphisms (e.g., rs3087243) associated with diseases such as systemic lupus erythematosus (SLE) and autoimmune thyroiditis. Genome-wide association studies (GWAS) have further illuminated the polygenic architecture of autoimmunity, identifying over 100 susceptibility loci for SLE as of 2025, many of which overlap with other autoimmune disorders and involve immune regulation pathways. These studies highlight shared genetic risks across diseases, explaining up to 50% of SLE . Epigenetic modifications, such as differences observed in twins discordant for SLE or , also modulate and contribute to disease discordance despite identical genomes, with hypomethylation in immune-related genes like IFI44L noted in affected twins. Autoimmunity typically follows a polygenic pattern, where multiple low-effect variants cumulatively increase risk rather than a single dominating. This is compounded by incomplete , as illustrated by , where only about 1-5% of carriers develop despite the allele's strong association. Such incomplete reflects interactions with other genetic and non-genetic factors in disease manifestation.

Endogenous Factors

Endogenous factors encompass internal physiological processes within the host that modulate the risk and progression of autoimmunity, independent of genetic inheritance or external exposures. These include sex-based differences, hormonal fluctuations, age-related changes, and metabolic states, each contributing to dysregulated immune responses through influences on immune cell function and tolerance mechanisms. Sex differences play a prominent role in autoimmunity, with females exhibiting a higher prevalence across most diseases, typically at a female-to-male ratio of approximately 4:1, and up to 9:1 or more in conditions like systemic lupus erythematosus (SLE). This disparity arises partly from X-chromosome dosage effects, where genes escaping inactivation in females can lead to overexpression of immune-related proteins, and the long non-coding RNA Xist, which coats the inactive X chromosome and has been shown to trigger innate immune sensing and autoantibody production in murine models of SLE. Additionally, estrogen enhances B-cell survival, activation, and autoantibody secretion, promoting humoral autoimmunity in females. Hormonal influences further shape autoimmune susceptibility, with dynamic changes across life stages affecting immune regulation. During , (RA) enters remission in 60-80% of cases, attributed to elevated levels of , progesterone, and , which expand regulatory T cells (Tregs) and shift cytokine profiles toward anti-inflammatory Th2 dominance. Conversely, marks an increased risk for autoimmune onset, particularly in females, as surging and levels post- drive pro-inflammatory immune shifts, including enhanced B- and T-cell responses that may initiate or exacerbate diseases like SLE and . Aging contributes to autoimmunity through progressive immune dysregulation, notably via inflammaging—a state of chronic, low-grade driven by accumulated senescent cells and persistent production, such as IL-6 and TNF-α. A key mechanism is , where the atrophies significantly by age 50, reducing output of naïve T cells and impairing central through defective negative selection of self-reactive clones and diminished Treg diversity, thereby elevating autoimmunity risk in older adults. Metabolic factors, such as , act as endogenous amplifiers of autoimmunity by altering adipose-derived signals. Excess adiposity induces a pro-inflammatory milieu through adipokines like , which is elevated in obesity and promotes Th1/Th17 polarization while suppressing Tregs, thereby facilitating disease progression in conditions like RA and SLE.

Environmental Triggers

Environmental triggers play a significant role in initiating or exacerbating autoimmune diseases by disrupting through mechanisms such as molecular , where microbial antigens resemble self-antigens, leading to cross-reactive immune responses. Infections are prominent examples; Epstein-Barr virus (EBV) infection has been strongly linked to (MS), with seroconversion increasing MS risk by 32-fold, often mediated by molecular between EBV nuclear antigen 1 and neuronal proteins like anoctamin 2. Similarly, group A Streptococcus infections trigger acute via molecular , where streptococcal antigens cross-react with cardiac and neuronal tissues, resulting in autoimmune-mediated valvular damage. Alterations in the gut microbiome, or , contribute to autoimmunity by impairing mucosal barrier function and promoting pro-inflammatory responses. In (IBD), is characterized by reduced microbial diversity, particularly a decline in Firmicutes phyla such as and Roseburia, which produce that maintain regulatory T cells and suppress . The hygiene hypothesis posits that reduced exposure to parasites and microbes in modern environments diminishes immune regulatory mechanisms, increasing susceptibility to both allergies and autoimmune conditions like and , as evidenced by protective effects of helminth in models. Chemical exposures and drugs can induce autoimmunity by altering immune cell function or promoting production. , an antiarrhythmic drug, is a classic trigger for , with 80-90% of long-term users developing antinuclear antibodies () over two years, though only a subset progress to clinical symptoms resembling systemic . Occupational exposure to silica dust is associated with systemic sclerosis (), particularly in patients, where the risk is increased up to 28-fold due to silica's adjuvant-like effects that enhance responses against antigens. Other environmental factors include (UV) radiation and . UV exposure, especially UVB, exacerbates systemic by inducing apoptotic cells that release autoantigens, triggering flares and photosensitive rashes in up to 70% of patients. elevates rheumatoid arthritis (RA) risk, particularly for anti-citrullinated protein antibody-positive disease, with ever-smokers facing approximately 1.7- to 2-fold increased odds compared to non-smokers, mediated by smoke-induced of proteins that breaks self-tolerance; this risk is amplified in genetically susceptible individuals carrying HLA-DRB1 shared alleles.

Pathophysiology

Initiation of Autoimmune Responses

The initiation of autoimmune responses begins with the breakdown of immunological tolerance, where self-antigens that are normally ignored or suppressed by regulatory mechanisms become targets of adaptive immune activation. This process often stems from failures in antigen presentation, where professional antigen-presenting cells (APCs) such as dendritic cells (DCs) fail to maintain tolerogenic signals, instead promoting proinflammatory responses. In particular, defects in DC maturation can lead to inadequate expression of costimulatory molecules or altered cytokine profiles, resulting in the priming of autoreactive T cells rather than their deletion or anergy. Such maturation defects have been linked to excessive DC activation in models of autoimmunity, exacerbating the loss of peripheral tolerance. A key stage in this initiation is the failure of proper , where self-peptides are displayed by (MHC) molecules in a context that drives effector T cell responses instead of . This can occur due to dysregulated loading or insufficient negative selection in the , allowing low-affinity autoreactive T cells to persist and expand upon encountering self-antigens in inflamed tissues. Once initiated, the response can broaden through epitope spreading, in which an initial immune reaction to a dominant self- releases additional sequestered antigens from damaged cells, leading to diversification of the autoreactive repertoire. This phenomenon has been observed in chronic inflammatory settings, where T and responses shift from a focused to a polyclonal attack on multiple epitopes. Infections play a pivotal role in triggering these events via bystander activation, where innate immune responses to pathogens—such as cytokine storms involving IL-6 and IL-23—nonspecifically activate nearby autoreactive lymphocytes without direct recognition. This mechanism allows self-reactive T cells to proliferate and infiltrate tissues during the inflammatory milieu created by . Complementing this, neoantigen formation arises from post-translational modifications of self-proteins, altering their to evade checkpoints; for instance, converts to in synovial proteins during (RA), generating novel epitopes that elicit anti-citrullinated protein antibodies (ACPAs). These modified antigens are particularly immunogenic in genetically susceptible individuals, bridging environmental insults to adaptive autoimmunity. At the cellular level, Th17 polarization emerges as a critical event in tissue-specific initiation, driven by TGF-β and IL-6 signaling that skews naïve + T cells toward IL-17 production, promoting recruitment and chronic . This polarization is amplified in barrier tissues like the gut or , where microbial signals intersect with self-antigen presentation to sustain autoreactivity. Experimental autoimmune (EAE), a mouse model mimicking (MS) initiation, illustrates these dynamics: immunization with (MOG) peptide leads to DC-mediated priming of Th17 cells, followed by blood-brain barrier breach and epitope spreading to other myelin antigens, recapitulating early autoimmunity.

Effector Mechanisms and Tissue Damage

In autoimmune diseases, once self-reactive lymphocytes escape mechanisms, effector arms of the drive pathological and tissue destruction. These processes involve coordinated actions of humoral and cellular immunity, often amplified by complement activation, leading to acute and chronic damage in affected organs. For instance, following the initiation of autoimmunity, persistent sustains these effectors, resulting in cycles of that culminate in and organ dysfunction. Humoral effector mechanisms primarily involve autoantibodies that form immune complexes with self-antigens, depositing in tissues and triggering reactions. In systemic (SLE), anti-nuclear antibodies bind to DNA and other nuclear components, forming complexes that deposit in glomerular basement membranes, activating local inflammation and causing . This process leads to and progressive renal failure if unchecked. Similarly, in rheumatoid arthritis (RA), autoantibodies target IgG, forming complexes in synovial tissues that exacerbate joint swelling and erosion. Cellular effectors, particularly T lymphocytes, play a central role in directing tissue-specific damage through release and direct . + T helper cells, differentiated into Th1 or Th17 subsets, secrete pro-inflammatory s such as interferon-gamma (IFN-γ) and interleukin-17 (IL-17), which recruit neutrophils and macrophages to inflamed sites. In , Th17 cells infiltrate synovial joints, where IL-17 promotes activation and degradation, contributing to bone erosion. + cytotoxic T cells, meanwhile, recognize autoantigens on target cells and induce via perforin and granzyme release; this is evident in , where + T cells destroy pancreatic beta cells, leading to insulin deficiency. Complement activation serves as a critical amplifier of both humoral and cellular effectors, enhancing opsonization and membrane attack complex formation that lyses target cells. In , autoantibodies against the at neuromuscular junctions activate the , depositing fragments and causing muscle weakness through synaptic destruction. This deposition not only recruits inflammatory cells but also perpetuates a feedback loop of autoantibody production. Complement inhibition has shown therapeutic promise in reducing such damage. Chronic effector activity culminates in irreversible tissue remodeling, including and organ failure, as repeated inflammatory insults replace functional with . In , persistent Th2 cytokine-driven activation leads to excessive deposition in skin and lungs, impairing organ function. Likewise, in , ongoing beta-cell destruction by CD8+ T cells and associated results in and complete loss of insulin production. These long-term effects underscore the need for early to halt effector progression.

Classification of Autoimmune Diseases

Organ-Specific vs. Systemic Autoimmunity

Autoimmune diseases are broadly classified into organ-specific and systemic categories based on the scope of immune-mediated damage. Organ-specific autoimmunity targets a single organ or tissue, leading to localized , whereas systemic autoimmunity involves multiple organs and tissues, resulting in widespread . This taxonomic framework aids in understanding disease patterns, though the distinction is not always absolute. In organ-specific autoimmune diseases, the is directed primarily against antigens in a particular organ, often driven by T cell-mediated mechanisms that infiltrate and destroy target tissues. For instance, mellitus involves autoreactive T cells attacking pancreatic beta cells, leading to insulin deficiency, while features T cell infiltration and subsequent of the gland. These conditions typically manifest with symptoms confined to the affected organ, such as hyperglycemia in or in Hashimoto's. The predominance of T cells in these diseases underscores their role in direct and within the target tissue. Systemic autoimmune diseases, by contrast, feature dysregulated immune responses that produce autoantibodies and immune complexes affecting diverse organs through vascular and involvement. Systemic lupus erythematosus (SLE), for example, targets skin, kidneys, joints, and other sites via anti-nuclear antibodies, causing multisystemic symptoms like , , and . Similarly, Sjögren's syndrome initially affects exocrine glands but often extends to extra-glandular sites such as lungs and , involving both T and hyperactivity. These disorders frequently require immunosuppressive therapies due to their diffuse impact. The boundary between organ-specific and systemic autoimmunity exists on a spectrum rather than as a strict , with some diseases exhibiting overlaps based on criteria such as the number of affected organ systems or the presence of circulating autoantibodies. For example, diseases initially classified as organ-specific may progress to involve additional sites, or vice versa, influenced by shared genetic and environmental factors. This continuum complicates precise categorization but highlights the interconnected nature of autoimmune processes. Systemic autoimmune diseases tend to be rarer and more severe than organ-specific ones, with higher morbidity due to multi-organ involvement. The annual incidence of SLE, a prototypical systemic disorder, is approximately 5.1 per 100,000 person-years globally, compared to higher rates for organ-specific conditions like (0.3–1.5 per 1,000 annually) or (15 per 100,000). Overall, autoimmune diseases affect approximately 5–10% of the global population, with organ-specific forms comprising the majority due to their higher prevalence in common targets like the and .

Major Autoimmune Disorders

Autoimmune disorders can be broadly categorized into organ-specific and systemic types, with numerous prominent examples illustrating the diversity of immune-mediated damage to targeted tissues.

Organ-Specific Autoimmune Disorders

Organ-specific autoimmune diseases primarily affect a single organ or tissue, leading to localized pathology. is characterized by immune-mediated demyelination in the , resulting in neurological deficits such as vision loss and motor impairment. involves autoantibodies against gastric parietal cells, impairing production and absorption, which causes . features stimulating autoantibodies against the receptor, leading to and symptoms like and . , encompassing conditions like , features immune attacks on the gut barrier, leading to chronic inflammation, ulceration, and complications such as strictures.

Systemic Autoimmune Disorders

Systemic autoimmune diseases involve widespread immune dysregulation affecting multiple organs. (RA) is marked by chronic synovial inflammation driven by autoantibodies such as and anti-citrullinated protein antibodies, causing joint erosion and deformity. , also known as , entails progressive fibrosis of connective tissues due to autoimmune activation, resulting in skin thickening and potential visceral involvement like . Recent reports from the 2020s have highlighted potential autoimmune links in , where post-viral infection with triggers the production of functional autoantibodies targeting neural and other tissues, contributing to persistent symptoms like and . Incidence trends for certain autoimmune disorders are rising; for instance, celiac disease prevalence doubled in from the late to around , with continued modest increases thereafter attributed to environmental and diagnostic factors.

Autoimmune Clusters

Autoimmune polyendocrine syndromes represent clusters of organ-specific autoimmunities affecting multiple endocrine glands. (APS-1), a rare caused by AIRE mutations, typically includes , , and chronic mucocutaneous due to T-cell dysregulation. (APS-2), more common and polygenic, combines with autoimmune or , often presenting in adulthood with overlapping endocrine failures.

Clinical Diagnosis

Diagnostic Criteria and Tests

Diagnosing autoimmune diseases relies on a combination of clinical evaluation and laboratory tests that detect autoantibodies, assess immune cell profiles, and visualize tissue damage, with an emphasis on tests offering high specificity to confirm autoimmunity. Autoantibody detection is a cornerstone, starting with screening assays like the antinuclear antibody (ANA) test, which exhibits high sensitivity of 95-99% for systemic lupus erythematosus (SLE) via indirect immunofluorescence on HEp-2 cells. More specific autoantibodies, such as anti-double-stranded DNA (anti-dsDNA) antibodies, provide diagnostic confirmation for SLE with specificities ranging from 90% to 98%, particularly when using assays like Crithidia luciliae immunofluorescence. In rheumatoid arthritis (RA), rheumatoid factor (RF) and anti-cyclic citrullinated peptide (anti-CCP) antibodies are key; anti-CCP demonstrates superior specificity (up to 98% when combined with RF positivity), aiding early diagnosis and distinguishing RA from other arthritides. Imaging and cellular analyses complement serological tests by providing direct evidence of immune-mediated damage. (MRI) is essential for (MS), where it identifies characteristic T2-hyperintense lesions in the brain and spinal cord, supporting the 2024 with over 90% sensitivity for demyelinating pathology and incorporating advanced features like the central vein sign for improved specificity. quantifies T-cell subsets, such as CD4+ helper and regulatory T cells, to evaluate imbalances in autoimmune conditions like SLE or , offering insights into disease activity through phenotypic profiling of naive, memory, and activated lymphocytes. Tissue biopsies provide definitive histopathological confirmation; for instance, in suspected reveals immune complex deposition and glomerular changes, guiding classification into stages I-VI per the International Society of Nephrology/Renal Pathology Society system. Standardized classification criteria integrate these tests with clinical features to achieve diagnostic certainty. The 2010 ACR/EULAR criteria for RA use a scoring system across joint involvement, serology (RF or anti-CCP), acute-phase reactants, and symptom duration, classifying RA if the total score reaches ≥6 out of 10 in patients with synovitis and no alternative diagnosis. For SLE, the 2019 EULAR/ACR criteria require an entry of ANA titer ≥1:80 by immunofluorescence, followed by a weighted score of ≥10 points across clinical domains (e.g., constitutional, hematologic, neuropsychiatric, mucocutaneous, serosal, musculoskeletal, renal; maximum 10 points) and immunologic domains (e.g., anti-dsDNA, anti-Smith; maximum 6 points), improving both sensitivity and specificity over prior criteria. Recent advances enhance diagnostic precision through technology integration. As of 2025, AI-assisted in ANA immunofluorescence, using systems like akiron® NEO, achieves moderate to very good agreement with manual interpretation, automating classification of HEp-2 cell patterns to reduce subjectivity and improve throughput in screening for diseases. Multiplex assays, such as bead-based Luminex platforms, simultaneously detect panels of autoantibodies (e.g., ANA, anti-CCP, anti-dsDNA) from a single sample, offering higher efficiency and specificity compared to traditional single-analyte ELISAs for broad autoimmune profiling.

Differential Diagnosis

Differentiating autoimmune diseases from their mimics is essential, as conditions such as and malignancies can present with overlapping clinical features, leading to diagnostic delays or errors. , particularly viral and bacterial, often simulate organ-specific autoimmunity through mechanisms like molecular mimicry, where microbial antigens trigger cross-reactive immune responses against self-tissues. Similarly, paraneoplastic syndromes associated with cancers can induce autoantibodies that mimic systemic autoimmune disorders, such as or , due to tumor-driven immune dysregulation. These syndromes occur in up to 10-20% of cancer patients and may feature autoantibodies like anti-Hu or anti-Yo, simulating or cerebellar degeneration; examples include mimicking undifferentiated with positive and . These mimics complicate evaluation, especially in early disease stages where symptoms like and predominate. Common infectious mimics include viral arthritis resembling (RA), where causes symmetric small-joint in up to 60% of cases, often with low-titer positivity. Bacterial infections like , caused by , can present with mimicking (MS), including , , and white matter lesions on MRI. For malignancies, paraneoplastic syndromes may feature autoantibodies like anti-Hu or anti-Yo, simulating or cerebellar degeneration; examples include mimicking undifferentiated with positive and . Diagnostic challenges arise from symptom overlap and serological pitfalls, such as (ANA) positivity in up to 20% of healthy individuals, which can lead to false positives without clinical correlation. , , and nonspecific inflammation further blur distinctions, particularly when infections lack fever or when cancers present with constitutional symptoms before overt tumor detection. Drug reactions versus drug-induced autoimmunity pose another hurdle; syndromes may cause rash and mimicking , but true drug-induced lupus erythematosus (DILE), affecting 15,000-30,000 annually and linked to drugs like , features antihistone antibodies and resolves upon discontinuation. Strategies for differentiation emphasize clinical patterns and targeted testing. Temporal progression aids distinction: acute onset with fever suggests , while chronic relapsing courses favor autoimmunity. Response to empiric antibiotics (e.g., improvement in within weeks) or lack of response to steroids can guide exclusion of infectious etiologies. For suspected paraneoplastic mimics, imaging like PET-CT and tumor marker screening are crucial, alongside autoantibody panels to identify onconeural antibodies. is valuable for monogenic mimics, such as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), which presents with multi-organ autoimmunity due to AIRE mutations and can be confirmed via sequencing to differentiate from polygenic forms. Illustrative cases highlight these issues. In Lyme disease versus MS, overlapping neurological symptoms like numbness and cognitive impairment occur, but Lyme typically follows tick exposure with erythema migrans rash in 70-80% of cases, confirmed by two-tier serology, whereas MS shows multifocal demyelination on MRI without infectious markers. For drug reactions versus DILE, a patient on procainamide developing arthralgia and positive ANA requires temporal association with drug initiation (symptoms after 1-3 months) and exclusion of hypersensitivity via skin biopsy, with DILE improving post-discontinuation unlike persistent idiopathic autoimmunity. These approaches underscore the need for multidisciplinary evaluation to avoid misattribution.

Management and Therapies

Pharmacological Interventions

Pharmacological interventions for autoimmunity primarily involve immunosuppressive agents, biologics, and targeted therapies designed to modulate dysregulated immune responses while minimizing off-target effects. These treatments vary by type, with organ-specific autoimmunity often requiring localized suppression and systemic conditions demanding broader immune modulation. Immunosuppressants form the cornerstone of therapy for many autoimmune diseases, providing rapid control of inflammation and flares. Corticosteroids, such as , are widely used for acute flares due to their potent and immunosuppressive effects, achieved by inhibiting pro-inflammatory production and modulating immune cell functions. In conditions like (RA) and systemic lupus erythematosus (SLE), prednisone doses of 5-60 mg daily effectively induce remission, though long-term use is tapered to avoid complications. Disease-modifying antirheumatic drugs (DMARDs), including , serve as first-line maintenance therapy for RA by inhibiting , thereby disrupting metabolism and reducing T-cell proliferation and release. Methotrexate, typically dosed at 7.5-25 mg weekly, achieves clinical remission in approximately 30-40% of RA patients when used early. Biologic therapies target specific immune components, offering precision over traditional immunosuppressants. Anti-tumor necrosis factor (TNF) agents, such as , are approved for and other inflammatory bowel diseases, where they neutralize TNF-α to halt cytokine-driven inflammation and promote mucosal healing. , administered intravenously at 5 mg/kg every 8 weeks after induction, induces remission in up to 60% of moderate-to-severe cases. Monoclonal antibodies like rituximab deplete B cells by binding , reducing production; it is effective in ANCA-associated , achieving remission in 60-70% of patients to standard . Rituximab dosing involves two 1 g infusions two weeks apart, with maintenance every 6 months. Targeted small-molecule therapies and cellular approaches address intracellular signaling pathways. Janus kinase (JAK) inhibitors, such as , block JAK-STAT signaling downstream of multiple cytokines (e.g., IL-6, IL-12), suppressing synovial inflammation in RA. , at 5-10 mg twice daily, improves disease activity scores in 50-70% of RA patients inadequately responsive to . Emerging chimeric antigen receptor (CAR) T-cell therapies, targeting on B cells, show promise for refractory SLE; phase 1/2 trials in the 2020s demonstrated complete remission in 70-100% of severe cases, with durable responses up to 2 years post-infusion. Common side effects of these interventions include heightened infection risk due to immune suppression, with serious occurring 2- to 5-fold more frequently in treated patients compared to the general . Corticosteroids and biologics particularly elevate to opportunistic pathogens like . Monitoring protocols involve baseline screening for latent (e.g., , ), regular complete blood counts, and prophylactic antibiotics or vaccinations as needed; for example, rituximab recipients require immunoglobulin level checks every 3-6 months. , including anti-drug antibody assays, guides dose adjustments to optimize efficacy and safety.

Lifestyle and Nutritional Approaches

Lifestyle modifications, including dietary changes and physical activity, play a supportive role in managing autoimmune diseases by potentially reducing and symptom severity. The , rich in fruits, vegetables, whole grains, fish, and olive oil, has been associated with decreased inflammatory activity in () patients. One demonstrated improvements in disease activity scores following a 12-week intervention compared to a control group. For celiac disease, a remains the cornerstone of management, effectively alleviating intestinal damage and symptoms by preventing immune-mediated responses to . supplementation is recommended for () patients, where deficiency is prevalent and linked to increased disease risk; typical doses range from 1000 to 4000 IU daily to maintain sufficient serum levels and potentially mitigate clinical activity. Regular exercise can alleviate and enhance overall well-being in autoimmune conditions. Moderate aerobic activity, such as walking or swimming for 150 minutes per week, has been shown to reduce in systemic lupus erythematosus (SLE) patients, with meta-analyses confirming improvements in physical function and . Mind-body practices like further support management by lowering stress levels, which are tied to elevations that may trigger flares; an eight-week program reduced pain and altered profiles in women with chronic pain conditions akin to autoimmune flares. Beyond diet and exercise, other behavioral interventions offer benefits. significantly lowers disease activity and cardiovascular risk in , with studies indicating that quitting leads to reduced progression compared to continued . Adequate , aiming for 7-9 hours nightly, helps regulate production and curb , as disruptions exacerbate pro-inflammatory responses in autoimmune disorders. Probiotics show promise for modulating and reducing in autoimmune diseases, but evidence remains inconsistent. Recent 2025 meta-analyses highlight modest improvements in disease activity and immune markers for conditions like and SLE, though results vary by strain and patient population, underscoring the need for further standardized trials.

References

  1. [1]
    Biochemistry, Autoimmunity - StatPearls - NCBI Bookshelf - NIH
    Dec 19, 2022 · Autoimmunity refers to an aberration in the body's normal development that causes the immune system to mount an attack against its cells.
  2. [2]
    Definition of Autoimmunity & Autoimmune Disease
    Autoimmunity is the presence of antibodies (which are made by B lymphocytes) and T lymphocytes directed against normal components of a person (autoantigens).
  3. [3]
    Autoimmune Diseases | NIAID
    Mar 20, 2025 · An estimated 8% of the U.S. population has an autoimmune disease. In this family of disorders, the immune system mistakenly attacks healthy, ...
  4. [4]
    Women and Autoimmune Diseases - Volume 10, Number 11 ... - CDC
    Autoimmune diseases affect approximately 8% of the population, 78% of whom are women. The reasons for the high prevalence in women are unknown, ...
  5. [5]
    Autoimmune disorders: MedlinePlus Medical Encyclopedia
    Jun 12, 2023 · An autoimmune disorder occurs when the body's immune system attacks and destroys healthy body tissue by mistake. There are more than 80 autoimmune disorders.Addison disease · Sjogren syndrome · Dermatomyositis · Myasthenia gravis
  6. [6]
    Prevalence of Autoimmune Diseases - Johns Hopkins Pathology
    These estimates suggest that all autoimmune diseases combined affect about 3% of the US population, thus approximately 10 million people.
  7. [7]
    The Increasing Prevalence of Autoimmunity and ... - PubMed Central
    Estimates of the yearly increases in the overall worldwide incidence and prevalence of autoimmune diseases are 19.1% and 12.5%, respectively [26]. Despite the ...
  8. [8]
    Understanding Autoimmunity: Mechanisms, Predisposing Factors ...
    Autoimmunity refers to an organism's immune response against its own healthy cells, tissues, or components, potentially leading to irreversible damage to vital ...
  9. [9]
    In brief: The innate and adaptive immune systems - NCBI - NIH
    Aug 14, 2023 · If the innate (general) immune system fails to destroy the germs, the adaptive (specialized) immune system takes over. The adaptive immune ...Protection offered by immune... · The adaptive immune system... · cells
  10. [10]
    Autoimmunity - an overview | ScienceDirect Topics
    Autoimmunity is defined as a phenomenon in which the immune response is directed against the body's own cells and tissues, leading to autoimmune disorders ...
  11. [11]
    Nature and functions of autoantibodies - PMC - PubMed Central
    Natural autoantibodies are mainly IgM, are encoded by unmutated V(D)J genes and display a moderate affinity for self-antigens. They provide a first line of ...
  12. [12]
    Lupus and other autoimmune diseases: Epidemiology in the ...
    The estimated prevalence of ADs outside Africa is 5% to 10%, with people of African ancestry being particularly affected.10, 11, 12 From 1965 (when ADs were ...Review Article · Epidemiology · Infectious Agents<|control11|><|separator|>
  13. [13]
    Evolution and Autoimmunity - NCBI - NIH
    Evolutionary theories can help us understand the origin of autommune diseases beginning with the concepts of genetic drift.Introduction · Evolutionary theories that may... · Influence of the first form of...
  14. [14]
    The Biological Significance of Evolution in Autoimmune Phenomena
    The human species have a cumulative evolution of the most complex mechanisms of innate and acquired immunity which makes it very vulnerable to autoimmunity.
  15. [15]
    Hashimoto Thyroiditis - StatPearls - NCBI Bookshelf
    Feb 9, 2025 · Hashimoto thyroiditis is an autoimmune disease that destroys thyroid follicular cells through cell- and antibody-mediated immune processes.
  16. [16]
    Rheumatoid arthritis - Autoimmunity - NCBI Bookshelf - NIH
    Persistent inflammation leads to destruction of cartilage and bone tissue with the consequent of misalignment and joint deformity (201). The involvement of RA ...
  17. [17]
    Serum immunoglobulin levels and naturally occurring antibodies ...
    Naturally occurring antibodies against PPC were found in the serum of conventional mice, but were severely reduced in GF-CD mice.
  18. [18]
    Microbiota and Autoimmunity - PMC - PubMed Central - NIH
    Autoimmune reactions can be both advanced or blocked by the commensal microbiota, which can affect innate and adaptive arms of immune responses.
  19. [19]
    A Threshold Model for T-Cell Activation in the Era of Checkpoint ...
    Mar 18, 2019 · As described here the Threshold Model provides a straightforward context for understanding immune activation, autoimmunity, and anti-tumor ...T Cell Exhaustion In Cancer · T Cell Response To Antigen... · Checkpoint Blockade...
  20. [20]
    Optimal T-cell receptor affinity for inducing autoimmunity - PNAS
    Nov 19, 2014 · Below-Threshold T Cells Comprise a Low Risk of Autoimmunity. We wondered how target cell antigen affinity influences the development of ...
  21. [21]
    Thyroidology over the ages - PMC - NIH
    Hippocrates and Gaius Plinius Secundus of Pliny regarded goiter to occur solely because of drinking of snow-water.[2,7,8] Initially goiter was not thought to be ...Missing: autoimmunity vitiligo
  22. [22]
    Croonian lecture.—On immunity with special reference to cell life
    1900Croonian lecture.—On immunity with special reference to cell lifeProc. ... Jennette J and Falk R (2010) The rise and fall of horror autotoxicus and forbidden ...
  23. [23]
    The 100th anniversary of Wassermann-Neisser-Bruck reaction
    The Wassermann reaction was used in the diagnosis of syphilis. The antigen used in it was prepared empirically. Originally, the so-called antigens were extracts ...
  24. [24]
    Autoimmune Hemolysis: A Journey through Time - PMC
    This paper will review the historical odyssey of how we have learned about the clinical manifestations, etiology, and management of AIHAMissing: 1894 | Show results with:1894
  25. [25]
    Hashimoto's Thyroiditis: Celebrating the Centennial Through ... - NIH
    It was initially described in 1912, but only rarely reported until the early 1950s. To celebrate this centennial, we reviewed the surgical pathology archives of ...Data Sources · Table 1 · Results
  26. [26]
    Paroxysmal cold hemoglobinuria - PubMed
    Paroxysmal cold hemoglobinuria is a rare cause of autoimmune hemolytic anemia predominantly seen as an acute form in young children after viral illnesses.Missing: 1910 | Show results with:1910
  27. [27]
    The Nobel Prize in Physiology or Medicine 1980 - Press release
    Dausset showed that within the HLA system in man there were two dominating regions, and Snell was subsequently able to show that this was the case, too, with ...Missing: autoimmunity | Show results with:autoimmunity
  28. [28]
    Mechanisms of central tolerance for B cells - Nature
    Apr 3, 2017 · Tolerance is regulated at the stage of immature B cell development (central tolerance) by clonal deletion, involving apoptosis, and by receptor editing.
  29. [29]
    The thymus and central tolerance - PubMed - NIH
    May 29, 2001 · This process of central tolerance (negative selection) reflects apoptosis and is a consequence of immature T cells receiving strong ...
  30. [30]
    What Do We Know About the Mechanisms of Elimination ... - PubMed
    This review describes current understanding of the mechanisms that mediate apoptosis of autoreactive lymphocytes during their development in primary lymphoid ...Missing: central | Show results with:central
  31. [31]
    Anergy into T regulatory cells: an integration of metabolic cues and ...
    Dec 17, 2018 · An intriguing relationship has emerged between two mechanisms of peripheral tolerance-induction of anergy and Foxp3 + regulatory T (Treg) cells- ...
  32. [32]
    Peripheral generation and function of CD4+CD25+ regulatory T cells
    Different types of regulatory T cells have been described, including CD4+CD25+ T cells that constitutively express CTLA-4, GITR and Foxp3, TGF-beta producing ...
  33. [33]
    The role of IL-10 and TGF-beta in the differentiation and effector ...
    Type 1 T regulatory (Tr1) cells are defined by their ability to produce high levels of IL-10 and TGF-beta, and these cytokines mediate their ability to suppress ...
  34. [34]
    CD28/B7 costimulation: a review - PubMed
    Finally, the CD28/B7 pathway could be involved with maintaining immune tolerance, as recent studies suggest the preferential binding of the B7-CTLA4 pathway ...
  35. [35]
    Mechanisms of peripheral T cell tolerance - PubMed
    Under these conditions, the T cells co-express Fas (CD95) and Fas ligand (FasL), and engagement of Fas triggers apoptotic death of the T cells. Mice with ...
  36. [36]
    Aire - PubMed - NIH
    Animal models of APECED have revealed that Aire plays an important role in T cell tolerance induction in the thymus, mainly by promoting ectopic expression of a ...
  37. [37]
    Common Variable Immunodeficiency and Autoimmune Diseases
    Jul 4, 2021 · Autoimmunity occurs in up to 30% of CVID patients (5–7), and it is frequently the presenting manifestation at the onset of immunodeficiency (1).
  38. [38]
    T Regulatory Cells in Primary Immune Deficiencies - PMC
    Defects in regulatory T cell generation and/or function are important contributors to autoimmunity in Primary immunodeficiency diseases · Defective generation of ...
  39. [39]
    Autoimmunity, immunodeficiency and mucosal infections: Chronic ...
    The most accepted explanation of how infection causes autoimmunity is based on the concept of “molecular mimicry” (similarity between the epitopes of an ...
  40. [40]
    Autoimmunity in Wiskott–Aldrich Syndrome: Updated Perspectives
    Aug 20, 2021 · Wiskott–Aldrich syndrome (WAS) is an uncommon X-linked combined-immunodeficiency disorder characterized by a triad of thrombocytopenia, eczema, ...
  41. [41]
    Serological Assessment for Celiac Disease in IgA Deficient Adults
    Selective immunoglobulin A deficiency is the most common primary immunodeficiency disorder that is strongly overrepresented among patients with celiac disease ( ...
  42. [42]
    Systemic lupus erythematosus in HIV: An insight into clinical ... - NIH
    Although HIV infection can mimic the manifestations of various autoimmune disorders, the coexistence of HIV and systemic lupus erythematosus (SLE) has been ...
  43. [43]
    Development of a secondary autoimmune disorder after ...
    Nov 21, 2006 · We studied the characteristics of affected patients, and the treatment and outcome of the secondary autoimmune disease to assess the factors ...
  44. [44]
    Twin concordance rates for rheumatoid arthritis - PubMed
    We conclude that the overall MZ concordance at 15% is lower than the 30% figure normally quoted from a study some 30 years ago and sets a ceiling at the ...
  45. [45]
    The British Isles survey of multiple sclerosis in twins - Neurology.org
    Including two suspected cases, 11 of 44 (25%) monozygotic twin pairs were concordant compared with two of 61 (3%) dizygotic twin pairs–two of 33 (6%) like-sexed ...
  46. [46]
    Genetics of rheumatoid arthritis: GWAS and beyond
    8,9 From such studies, the overall heritability of RA has been estimated to be between 50% and 60%.10. It is documented that genetic risk factors contribute to ...<|separator|>
  47. [47]
    Predominance of HLA-DRB1*0405 in Korean patients with ...
    RESULTS--The phenotype frequency of HLA-DR4 in RA patients was significantly greater than that in controls (60.0% versus 31.4%, odds ratio (OR) 3.28, 95% ...
  48. [48]
    Fifty years after the discovery of the association of HLA B27 ... - NIH
    Aug 31, 2023 · HLA B27 explains less than 30% of the total genetic load. About 60%–90% of axSpA patients worldwide carry HLA B27.
  49. [49]
    Autoimmunity-Related Risk Variants in PTPN22 and CTLA4 Are ...
    Apr 8, 2020 · Both, the PTPN22 rs2476601 and CTLA4 rs3087243 SNPs were shown to be associated with numerous autoimmune diseases, including T1D, SLE, and RA (8 ...
  50. [50]
    Multi-ancestry and multi-trait genome-wide association meta ...
    Feb 7, 2023 · GWAS have also been successful in SLE. To date, GWAS have identified >130 loci that reach genome-wide significance (P value <5 × 10−8) ...
  51. [51]
  52. [52]
    Epigenomics of autoimmune diseases - PMC - PubMed Central
    Furthermore, MZ twins discordant for SLE display differences in DNA methylation and expression of a number of genes associated with immune function. Transcript ...
  53. [53]
    Introduction to Genetics of Autoimmune Diseases - NCBI - NIH
    Polygenic determination refers to the mathematical model in which a number of genes with small additive effects provide an underlying genetic predisposition to ...
  54. [54]
    How common is ankylosing spondylitis? - MedicalNewsToday
    Jan 26, 2023 · Around 5–6%Trusted Source of people with the HLA-B27 gene have AS. While this percentage may appear quite low, it is many times higher than the ...Prevalence · Who AS affects · Males vs. females · Rates by countryMissing: penetrance | Show results with:penetrance
  55. [55]
  56. [56]
    Altered X-chromosome inactivation predisposes to autoimmunity
    May 3, 2024 · Males are globally more sensitive to infectious diseases, while females are more susceptible to systemic autoimmunity. X-chromosome inactivation ...Altered X-Chromosome... · Results · Perturbation Of Xci Leads To...<|separator|>
  57. [57]
    Rheumatoid arthritis and pregnancy: evolution of disease activity ...
    Sep 2, 2011 · Klipple and Cecere [5] reported ∼70% of patients improving during pregnancy, with >90% of them experiencing a disease relapse in the first year ...Abstract · Effect of pregnancy on disease... · Drug treatment of RA during...
  58. [58]
    Autoimmune Disease in Women: Endocrine Transition and Risk ...
    Prolactin is another pro-inflammatory hormone implicated in development of autoimmune diseases due to its increased concentrations found in post-pubertal ...
  59. [59]
    Contributions of Age-Related Thymic Involution to ... - PubMed Central
    Jan 20, 2020 · We discuss the research progress uncovering how age-related thymic involution contributes to immunosenescence and inflammaging, as well as their intersection.
  60. [60]
  61. [61]
    Epstein-Barr Virus in Multiple Sclerosis: Past, Present, and Future
    Sep 15, 2025 · EBV seroconversion was found to increase the risk of MS development by 32-fold, with a median of 5 years before MS diagnosis. These findings ...
  62. [62]
    Molecular mimicry between Anoctamin 2 and Epstein-Barr virus ...
    Aug 20, 2019 · Molecular mimicry between Anoctamin 2 and Epstein-Barr virus nuclear antigen 1 associates with multiple sclerosis risk. Proc Natl Acad Sci ...
  63. [63]
    Rheumatic Fever, Autoimmunity and Molecular Mimicry - NIH
    Studies have for some time supported the hypothesis that molecular mimicry between the group A streptococcus and heart was important in the immune responses in ...
  64. [64]
    The gut microbiota and inflammatory bowel disease - PMC - NIH
    The reduced diversity of the gut microbiota observed in IBD patients is largely due to a decline in the diversity of Firmicutes. Among Firmicutes, a decrease ...
  65. [65]
    The 'hygiene hypothesis' for autoimmune and allergic diseases - NIH
    Conversely, infection of NOD mice with a wide variety of bacteria, virus and parasites protects completely ('clean' NOD mice) from diabetes [2]. Similarly ...
  66. [66]
    Drug-Induced Glomerular Disease: Immune-Mediated Injury - PMC
    Jun 19, 2015 · Here, we review drug-induced, ANCA-associated vasculitis (AAV), drug-induced lupus (DIL), and drug-associated membranous nephropathy (MN).
  67. [67]
    Clinical characteristics of systemic sclerosis patients with ... - NIH
    Sep 15, 2023 · Increasing evidence shows that SSc is related to various occupational and environmental factors, such as silica exposure [2–4]. The risk of SSc ...Missing: odds | Show results with:odds
  68. [68]
    Association of Ultraviolet-B Radiation and Risk of SLE among ... - NIH
    Jul 1, 2023 · Ultraviolet radiation (UV) exposure is associated with photosensitivity, rashes, and flares in systemic lupus erythematosus (SLE).
  69. [69]
    Quantification of the influence of cigarette smoking on rheumatoid ...
    Results: Current smokers, ex-smokers, and ever-smokers of both sexes had an increased risk for seropositive RA (for ever-smokers the odds ratio was 1.7 (95% ...<|control11|><|separator|>
  70. [70]
    The role of dendritic cells in autoimmunity - PMC - PubMed Central
    In this Review, we discuss the proposed roles of DCs in immunological tolerance, the effect of the gain or loss of DCs on autoimmunity and DC-intrinsic ...Missing: defects | Show results with:defects
  71. [71]
    Molecular Control of Steady-State Dendritic Cell Maturation and ...
    Dcir deficiency causes development of autoimmune disease in mice due to excess expansion of dendritic cells. Nat Med. 2008;14(2):176–80. doi: 10.1038/nm1697 ...
  72. [72]
    Antigen Presentation, Autoantigens, and Immune Regulation in ...
    Antigen presentation is in the center of every autoimmune disease, influencing both tolerance induction in the thymus and self-antigen recognition and ...
  73. [73]
    Epitope spreading - PMC - PubMed Central - NIH
    Epitope (determinant) spreading is the development of immune responses to endogenous epitopes secondary to the release of self antigens during a chronic ...
  74. [74]
    Emerging role of bystander T cell activation in autoimmune diseases
    During an immune response to a pathogen, bystander activation of self-reactive T cells via inflammatory mediators such as cytokines can trigger autoimmune ...
  75. [75]
    Post‐Translational Modified Neoantigens in Autoimmune Diseases
    Individuals carrying the HLA‐DR4 allele are more susceptible to RA when exposed to smoke, which induces citrullination of proteins and produces neoantigens ...
  76. [76]
    Th17 cell pathogenicity in autoimmune disease - PMC
    Sep 1, 2025 · Here we discuss the pathogenic roles of Th17 cells in autoimmune diseases such as multiple sclerosis, inflammatory bowel disease and psoriasis, ...
  77. [77]
    Experimental autoimmune encephalomyelitis (EAE) as a model for ...
    Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple ...
  78. [78]
    Classification of Autoimmune Diseases - Johns Hopkins Pathology
    Systemic autoimmune diseases are those where the autoantigens are found in almost any type of cell in the body, for example the DNA - protein complexes.<|control11|><|separator|>
  79. [79]
    Autoimmune responses are directed against self antigens - NCBI - NIH
    Autoimmune diseases can be classified into clusters that are typically either organ-specific or systemic. The classification of disease is an uncertain ...
  80. [80]
    Untangling a Complex Web: How to Categorize Autoimmune Disease
    In organ-specific autoimmune diseases, a person's immune system attacks a single system, organ, or tissue. Examples include Graves' disease in the thyroid gland ...
  81. [81]
    Helper T cells in antibody-mediated, organ-specific autoimmunity
    The production of pathogenic autoantibodies in organ-specific autoimmune diseases is largely T cell dependent. For many of these diseases, the precise ...
  82. [82]
    T cells in the control of organ-specific autoimmunity - PMC
    Autoimmune diseases result from an imbalance of pathogenic autoreactive Tconv cells and protective Tregs. Many immunotherapies for autoimmune diseases share a ...
  83. [83]
    Autoimmune disorders | Better Health Channel
    Autoimmune disorders are broadly grouped into two categories – 'organ-specific' means one organ is affected, while in 'non-organ-specific' disorders, multiple ...Summary · On This Page · Types Of Autoimmune...Missing: classification | Show results with:classification
  84. [84]
    Systemic Autoimmune Disease - an overview | ScienceDirect Topics
    Autoimmune diseases are classified into two main groups of organ-specific autoimmune diseases and systemic autoimmune diseases. The first one is indicative ...<|control11|><|separator|>
  85. [85]
    Immunogenetic mechanisms for the coexistence of organ-specific ...
    Organ-specific autoimmune diseases affect particular targets in the body, whereas systemic diseases engage multiple organs. Both types of autoimmune diseases ...Missing: spectrum | Show results with:spectrum
  86. [86]
    The global epidemiology of SLE: narrowing the knowledge gaps - NIH
    Mar 29, 2023 · Overall incidence was estimated at 5.1 (95% CI 4.6, 5.6) per 100 000 person-years [3], with increased incidence rates in Black females (15.9), ...
  87. [87]
    Hashimoto Thyroiditis - DynaMed
    May 3, 2024 · Up to 2% of the general population is reported to have Hashimoto thyroiditis. 3. The reported annual incidence is 0.3-1.5 cases per 1,000 ...
  88. [88]
    Prevalence and incidence of type 1 diabetes in the world
    The results of meta-analysis showed that the incidence of type 1 diabetes was 15 per 100,000 people and the prevalence was 9.5% (95% CI: 0.07 to 0.12) in the ...
  89. [89]
    Human autoimmune diseases: a comprehensive update - Wang
    Jul 25, 2015 · Historically, autoimmune diseases were considered to be rare but, through rigorous epidemiological studies, have now been shown to affect 3–5% ...<|control11|><|separator|>
  90. [90]
    A causal link between autoantibodies and neurological symptoms in ...
    Jun 19, 2024 · Acute SARS-CoV-2 infection triggers the generation of diverse and functional autoantibodies (AABs), even after mild cases.
  91. [91]
    The Role of Environmental Factors in the Development of Celiac ...
    The total prevalence of celiac disease has, indeed, doubled in Finland during the last two decades (1.05% in 1978–1980 and 1.99% in 2000–2001) [9], and the ...<|control11|><|separator|>
  92. [92]
    Autoimmune Polyglandular Syndromes - Endotext - NCBI Bookshelf
    Jul 21, 2024 · APS2 is characterized by the triad of T1DM, Addison disease, and thyroid autoimmunity with hypothyroidism, hyperthyroidism, or Hashimoto ...ABSTRACT · ADRENAL INSUFFICIENCY · AUTO-IMMUNITY...
  93. [93]
    Anti-dsDNA Testing Specificity for Systemic Lupus Erythematosus
    Jan 5, 2022 · Anti-dsDNA testing shows considerable variation in test specificity, with potential impact on the management of SLE patients.
  94. [94]
    Magnetic Resonance Imaging (MRI) for Diagnosing Multiple Sclerosis
    The number of lesions on an initial MRI can help you assess your risk of developing a second attack in the future and being diagnosed with MS. The MRI can also ...
  95. [95]
    TCP - Overview: T-Cell Subsets, Naive, Memory, and Activated, Blood
    Determining the presence of naive, memory, and activated T cells in various clinical contexts including autoimmune diseases, immunodeficiency states.
  96. [96]
    2010 Rheumatoid arthritis classification criteria: an American ...
    To classify a patient as having or not having definite RA, a history of symptom duration, a thorough joint evaluation, and at least one serological test (RF or ...
  97. [97]
    Pilot Study of AI-Assisted ANA Immunofluorescence Reading ...
    Pilot Study of AI-Assisted ANA Immunofluorescence Reading-Comparison with Classical Visual Interpretation. J Clin Med. 2025 Sep 30;14(19):6924.
  98. [98]
    Is It Rheumatoid Arthritis or an Infectious Disease Mimic?
    Feb 18, 2022 · Rheumatoid arthritis closely mimics various mycobacterial, bacterial, and viral infections, especially during the early stages of disease.
  99. [99]
    A case series of malignancies: mimics of Rheumatological disorder
    Malignancies can present as autoimmune diseases or they can mimic flare of Rheumatological diseases. Cancer can present as vasculitides, CTDs, myositis or ...
  100. [100]
    Lyme Disease Vs. Multiple Sclerosis - WebMD
    Jan 31, 2025 · Multiple sclerosis (MS) and Lyme disease are diseases that stem from very different causes. MS is a life-long autoimmune condition that disrupts the ...
  101. [101]
    Misdiagnosing Lyme disease: The Great Imitator
    Patients with Lyme disease have been incorrectly diagnosed with: multiple sclerosis (MS), fibromyalgia, chronic fatigue syndrome, autoimmune diseases.
  102. [102]
    Paraneoplastic and Other Autoimmune Disorders of the Central ...
    Autoimmune CNS disorders may be paraneoplastic or idiopathic in etiology. Improvements in existing methodologies and new laboratory techniques for detecting ...Missing: differential | Show results with:differential
  103. [103]
    Immune phenotypes in individuals positive for antinuclear antibodies
    Indeed, as many as 20% of the otherwise healthy individuals can express an ANA as detected by the usual serological assays.
  104. [104]
    Drug-Induced Autoimmune Diseases - Pharmacy Times
    Jan 20, 2016 · When making a diagnosis, a clinician must be aware of the differential diagnosis, which includes drug hypersensitivity, eosinophilia ...
  105. [105]
    Monogenic Autoimmune Diseases: Insights into Self-Tolerance - PMC
    An example of a rare monogenic autoimmune diseases is autoimmune polyendocrinopathy syndrome (APS) type I (OMIM 240300), also known as autoimmune ...
  106. [106]
    Treating Human Autoimmunity: Current Practice and Future Prospects
    Traditional therapies for autoimmune disease have relied on immunosuppressive medications that globally dampen immune responses. These agents are highly ...
  107. [107]
    Targeted Immunotherapy for Autoimmune Disease - PMC
    Advances in targeted immunotherapy during the past few decades have revolutionized the treatment and clinical outcomes of patients with autoimmune diseases.
  108. [108]
    Pharmacokinetics and Pharmacodynamics of Systemic ... - NIH
    Sep 12, 2024 · Corticosteroids are widely used in various autoimmune and inflammatory diseases due to their anti-inflammatory and immunosuppressive properties.
  109. [109]
    Systemic lupus erythematosus and glucocorticoids: A never-ending ...
    The three main basic principles are a) using methyl-prednisolone pulses to induce remission not only in severe disease; b) limiting initial doses of prednisone ...Abstract · Introduction · Activating Genomic And...
  110. [110]
    How does methotrexate work? - PubMed
    Apr 29, 2020 · In particular, the long-held dogma that this disease-modifying anti-rheumatic drug (DMARD) acts via the folate pathway does not appear to hold ...<|separator|>
  111. [111]
    [PDF] News from the IBD Help Center - BIOLOGICS - Fact Sheet
    Infliximab (Remicade®) has been approved for the treatment and maintenance of remission of moderate to severe Crohn's disease and ulcerative colitis (in adults ...
  112. [112]
    Discontinuation of Infliximab Therapy in Patients with Crohn's Disease
    Jun 14, 2022 · Tumor necrosis factor-α (TNFα) inhibitor therapy, such as infliximab, has improved the symptomatic and endoscopic control of Crohn's disease by ...
  113. [113]
    The effect of continuous B cell depletion with rituximab on ... - NIH
    Rituximab has become an important component of the treatment of antineutrophil cytoplasmic antibody (ANCA) vasculitis (2, 8, 9). The Rituximab in ANCA- ...
  114. [114]
    B-cell treatment in ANCA-associated vasculitis - PubMed
    May 1, 2020 · Rituximab (RTX), an anti-CD20 monoclonal antibody that targets B-cells, has proven its efficacy for induction of remission in severe ANCA vasculitis.
  115. [115]
    Tofacitinib Suppresses Several JAK-STAT Pathways in Rheumatoid ...
    Sep 24, 2021 · Tofacitinib suppresses multiple JAK-STAT pathways in cytokine and cell population specific manner in RA patients in vivo.
  116. [116]
    Tofacitinib Suppresses Several JAK-STAT Pathways in Rheumatoid ...
    Sep 23, 2021 · Tofacitinib has been shown to inhibit cytokine signaling and the effector functions of different immune cells and synovial fibroblasts in vitro ...
  117. [117]
    Allogeneic CD19-targeted CAR-T therapy in refractory systemic ...
    May 29, 2025 · In patients with severe and refractory SLE, allogeneic CAR-T cell therapy showed profound safety and clinical efficacy for disease remission.Missing: 2020s | Show results with:2020s
  118. [118]
    Overall and comparative safety of biologic and immunosuppressive ...
    Most consistent risk factors for serious infections include use of combination therapy with immunosuppressive agents and/or corticosteroids, moderate to severe ...
  119. [119]
    Screening and Monitoring for Infectious Complications When ...
    An approach for screening and monitoring of immunosuppression-associated infections for prospective use in clinical trials.
  120. [120]
    Therapeutic Drug Monitoring: A Tool to Optimize Treatment of ...
    Nov 20, 2023 · Therapeutic drug monitoring (TDM), which is individualized dosing based on assessment of drug levels and, when indicated, anti-drug antibodies, ...