Fact-checked by Grok 2 weeks ago

Histocompatibility

Histocompatibility refers to the immunological compatibility between tissues or organs of different individuals, primarily governed by the similarity of antigens encoded by the , a group of genes that produce cell-surface proteins critical for and regulation. In humans, the MHC is termed the system, located on and comprising over 200 genes that exhibit extreme polymorphism, ensuring diverse immune recognition capabilities across populations. MHC class I proteins are expressed on nearly all nucleated cells, while class II proteins are primarily expressed on antigen-presenting cells; these proteins play a pivotal role in distinguishing self from non-self antigens, thereby preventing autoimmune reactions while enabling targeted responses to pathogens. The MHC is divided into three classes, with class I (HLA-A, HLA-B, ) and class II (, HLA-DQ, HLA-DP) being the most relevant for histocompatibility; class I molecules present intracellular peptides, such as those from viruses or tumors, to cytotoxic + T cells, while class II molecules display extracellular antigens from engulfed pathogens to helper + T cells. This process, known as , allows T cells to recognize and respond specifically to peptide-MHC complexes, forming the foundation of adaptive immunity. In transplantation medicine, histocompatibility testing assesses HLA matching between donors and recipients to minimize graft rejection, as mismatches trigger alloreactive T-cell responses that can lead to acute or chronic organ failure. Beyond transplants, MHC variations influence susceptibility to autoimmune diseases, infections, and cancer, highlighting the complex interplay between and .

Fundamentals

Definition and Biological Importance

Histocompatibility refers to the extent to which tissues or organs from a donor and recipient share similar antigens, particularly cell surface proteins, that the recipient's recognizes as "self" or "non-self," thereby determining compatibility for transplantation. This compatibility is primarily governed by genetically encoded molecules known as histocompatibility antigens, which include major and minor variants, enabling the to distinguish between endogenous and exogenous entities. Biologically, histocompatibility is central to adaptive immunity, as these antigens, especially those from the (MHC), facilitate the presentation of peptide fragments from pathogens or abnormal cells to T lymphocytes. MHC molecules bind and display these peptides on cell surfaces, allowing T cells to initiate targeted immune responses against foreign invaders while tolerating self-tissues; this MHC restriction ensures precise recognition and prevents . Without sufficient histocompatibility, the recipient's T cells and antibodies mount aggressive responses against donor tissues, leading to through mechanisms such as hyperacute antibody-mediated damage, acute cellular rejection, or chronic . In practice, syngeneic transplants, where donor and recipient are genetically identical (e.g., identical twins), exhibit perfect histocompatibility and minimal rejection risk, serving as an ideal benchmark for . Conversely, allogeneic transplants from unrelated donors frequently involve histocompatibility mismatches, necessitating immunosuppressive therapies to mitigate T-cell activation and production against donor antigens. The phenomenon of histocompatibility was first observed in skin grafts exchanged between inbred strains of mice in the late , revealing the genetic basis of immune surveillance and tissue rejection.

Types of Histocompatibility Antigens

Histocompatibility antigens are broadly classified into types based on their genetic origins, polymorphism, and capacity to elicit immune responses. Major histocompatibility antigens are encoded by genes within the (MHC), known as human leukocyte antigens (HLA) in humans, and are characterized by their high degree of polymorphism, which enables them to present a diverse array of peptides to T cells and trigger robust immune reactions. The major antigens are divided into class I and class II molecules. Class I antigens, encoded by the , HLA-B, and genes, are expressed on nearly all nucleated cells and primarily present endogenous peptides to cytotoxic + T cells, facilitating the recognition and elimination of infected or abnormal cells. Class II antigens, encoded by the , HLA-DQ, and HLA-DP gene pairs, are predominantly expressed on antigen-presenting cells such as dendritic cells, macrophages, and B cells, where they display exogenous peptides to helper CD4+ T cells, thereby orchestrating adaptive immune responses including antibody production and T cell activation. In contrast, minor histocompatibility antigens are not encoded by MHC genes but arise from polymorphic peptides derived from endogenous proteins, which are presented by MHC molecules; these antigens exhibit lower polymorphism compared to major ones and provoke weaker, more delayed immune responses. A well-known example is the H-Y antigen, encoded by genes on the , which can elicit immune reactions in females receiving male donor tissues due to the absence of homologous X-chromosome variants. The relative potency of these antigens significantly influences transplant outcomes: mismatches in major histocompatibility antigens lead to rapid, acute rejection within days due to vigorous T cell activation, whereas minor antigen mismatches typically contribute to chronic complications, such as (GVHD), particularly in MHC-matched settings where they become the primary targets of donor T cells. The extreme polymorphism of major antigens is underscored by the identification of over 42,000 HLA alleles in the IPD-IMGT/HLA Database as of September 2025, reflecting ongoing that complicates matching in clinical transplantation.

Historical Development

Early Observations and Experiments

In , pioneering experiments with inbred mouse strains laid the groundwork for understanding histocompatibility. Clarence C. Little and Arthur M. Cloudman utilized these genetically uniform strains, developed through successive generations of brother-sister matings at the Roscoe B. Jackson Memorial Laboratory, to investigate tumor transplantation. Their studies demonstrated that grafts were readily accepted between identical siblings from the same inbred line but consistently rejected when transplanted between non-identical siblings from different strains, indicating that genetic differences controlled . This observation highlighted the role of multiple heritable factors—termed "rejection genes"—in mediating graft outcomes, providing the first of histocompatibility barriers in mammals. Building on these findings, Peter B. Medawar conducted seminal studies in the that elucidated the immunological basis of graft rejection. During , Medawar developed techniques for in rabbits to treat burn victims, observing that homografts from unrelated donors were invariably rejected after an initial period of acceptance, while autografts persisted indefinitely. Extending this to mice using inbred strains, he demonstrated that rejection involved an adaptive immune response, as prior exposure to donor tissue accelerated subsequent graft destruction—a phenomenon termed the "second-set" reaction. For these contributions, which established rejection as an antigen-specific process rather than mere mechanical failure, Medawar shared the 1960 in or with Frank Macfarlane Burnet for discoveries concerning immunological . In the 1950s, Medawar, along with Rupert Billingham and , advanced these insights through experiments on acquired immunological tolerance. They showed that injecting foreign cells into neonatal mice prior to immune system maturation induced lifelong acceptance of skin grafts from the same donor strain, without compromising responses to unrelated antigens. This prenatal or neonatal exposure effectively "trained" the to recognize the foreign tissue as self, marking a breakthrough in understanding how tolerance could be artificially induced to prevent rejection. These mouse-based findings directly informed early human applications. Early clinical observations in humans reinforced the genetic underpinnings of histocompatibility. In 1954, surgeons at Hospital, led by Joseph E. Murray, performed the first successful kidney transplant between identical twins, Ronald and Richard Herrick, where the graft functioned for over eight years without , underscoring the critical role of genetic identity in averting rejection. This milestone paved the way for subsequent research into the molecular basis of histocompatibility, culminating in the identification of the .

Establishment of MHC Framework

The establishment of the (MHC) framework in the mid-20th century built upon initial observations of by integrating genetic, serological, and cellular approaches to identify and characterize the key histocompatibility systems in mice and humans. In the 1960s, George D. Snell advanced the understanding of the mouse H-2 complex, a genetic region controlling strong histocompatibility responses, through extensive breeding of congenic strains that isolated H-2 as the primary locus influencing graft survival. Paralleling this, the (HLA) system emerged from serological studies; Jean Dausset's 1952 observation of leukocyte-agglutinating antibodies in sera from polytransfused patients marked the first observation of such antibodies, which led to the detection of human histocompatibility antigens, with the first specific (MAC, now HLA-A2) identified in 1958. Independently, Jon J. van Rood contributed to the early delineation of HLA specificities in the late 1950s and 1960s by identifying additional alloantibodies and their role in transfusion reactions and graft compatibility, facilitating the recognition of HLA as a polymorphic complex analogous to H-2. Key milestones in the 1960s and 1970s solidified the MHC framework through targeted mapping and functional assays. The first HLA-A and HLA-B loci were serologically defined and provisionally mapped during the Third International Histocompatibility Workshop in 1967, with formal nomenclature established at the 1968 World Health Organization (WHO) meeting, confirming their close genetic linkage and distinguishing them as class I antigens. In the 1970s, distinctions between MHC class I and class II molecules were clarified using serology for class I (expressed on most nucleated cells) and mixed lymphocyte reactions (MLR) for class II (primarily on antigen-presenting cells), where MLR identified HLA-D loci as the human equivalents of mouse I-region genes controlling T-cell proliferation. These assays revealed that class II disparities drove stronger proliferative responses in MLR, bridging serological typing with cellular immunity. The development of standardized nomenclature by the WHO HLA Nomenclature Committee, formed in 1968, provided a systematic framework for naming antigens and later alleles, evolving from broad serological designations (e.g., HLA-A1) in the to more precise allele-level resolution (e.g., HLA-A*02:01) as molecular techniques emerged in the 1980s and beyond. This enabled international collaboration and consistent reporting of HLA variants, essential for advancing genetic studies of the MHC. The foundational contributions of Snell, Dausset, and —who elucidated genes linked to MHC—were recognized with the 1980 in or for discoveries on surface structures regulating immune reactions.

Major Histocompatibility Complex

Genetic Organization and Polymorphism

The human (MHC), designated as the (HLA) system, is situated on the short arm of at locus 6p21.3, encompassing a genomic region of approximately 4 megabases (Mb) that ranks as the densest cluster of genes in the . This region is subdivided into three primary classes: class I, class II, and class III. The class I region, spanning about 1.8 Mb near the telomeric end, encodes classical HLA molecules , , and , alongside non-classical variants , , and , which contribute to on cell surfaces. The class II region, located centromerically to class I and covering roughly 0.9 Mb, includes the heterodimeric genes (with multiple DRB loci), HLA-DQ, and HLA-DP, essential for presenting extracellular antigens to T cells. In contrast, the class III region, positioned between classes I and II and spanning about 0.7 Mb, harbors over 50 genes unrelated to direct , such as those encoding complement components (e.g., C2, C4, factor B), cytokines (e.g., ), and heat shock proteins. Overall, the MHC contains more than 200 protein-coding genes, with its compact organization facilitating coordinated immune regulation. Polymorphism within the MHC is exceptionally high, representing the greatest density of genetic variants observed across vertebrate genomes, which enables diverse immune responses to pathogens. This variability is predominantly concentrated in the peptide-binding regions of classical HLA genes: for class I molecules, polymorphisms cluster in exons 2 and 3 encoding the α1 and α2 domains that form the -binding groove; for class II, they localize to exon 2 of the β-chain genes, shaping the antigen-binding cleft. Key loci such as , -B, -C, -DRB1, -DQB1, and -DPB1 exhibit thousands of alleles, with the majority of sequence differences altering peptide specificity and affinity. As of October 2025 (release 3.62), the authoritative IPD-IMGT/HLA Database catalogs 42,996 distinct HLA alleles, including 29,475 for class I and 13,521 for class II, underscoring the ongoing discovery of novel variants through next-generation sequencing. This extreme polymorphism is maintained by balancing selection, particularly from pathogen-driven pressures that favor heterozygous individuals capable of presenting a broader array of antigens, as evidenced by signatures of positive selection in binding-site codons across populations. MHC alleles are inherited as haplotypes—contiguous blocks of linked genes transmitted en bloc from parents—due to suppressed recombination within the region, resulting in pronounced (LD) that extends over megabases. For instance, the ancestral haplotype A1-B8-DR3, common in European-descended populations, demonstrates near-complete conservation across class I, II, and III loci, with LD values often exceeding 0.9, which influences disease associations and transplant compatibility. Such haplotype structures preserve co-evolved gene combinations, amplifying the functional impact of polymorphism while limiting independent assortment.

Mechanisms of Antigen Presentation

Antigen presentation by (MHC) molecules is a central process in adaptive immunity, enabling T cells to recognize foreign or altered-self peptides displayed on surfaces. In the classical pathways, molecules present endogenous antigens to + T cells, while molecules present exogenous antigens to + T cells, ensuring compartmentalized immune surveillance. These mechanisms rely on intricate intracellular trafficking and processing steps that generate peptide-MHC complexes capable of activating T cell receptors with high specificity. The antigen presentation pathway primarily handles endogenous antigens, such as viral proteins synthesized within the of infected cells. These proteins are degraded by the into short peptides, which are then transported into the (ER) via the transporter associated with antigen processing (TAP). In the ER, peptides are loaded onto nascent molecules, stabilized by chaperones like and tapasin, before the complex traffics to the cell surface for recognition by cytotoxic + T cells. This pathway ensures that intracellular threats, including viruses and tumors, are flagged for immune elimination. In contrast, the MHC class II pathway processes exogenous antigens acquired by or , typically by professional antigen-presenting cells like dendritic cells and macrophages. These antigens are delivered to lysosome-like compartments where they are degraded by acid hydrolases and cathepsins into peptides. molecules, assembled in the with the aid of the invariant chain (), traffic through the Golgi to these compartments (MHC class II compartments, or MIICs), where Ii is proteolytically removed, allowing peptide loading facilitated by . The resulting peptide- complexes are then transported to the plasma membrane to engage helper + T cells, orchestrating humoral and cellular responses. Peptide binding to MHC molecules occurs within polymorphic grooves formed by the α-helices of the MHC domains, dictating specificity and alloreactivity. For , the closed-ended groove accommodates peptides of 8-10 , anchored at both termini by conserved residues that interact with pockets in the groove. grooves are open-ended, binding longer peptides of 13-25 , with core regions of 9 residues fitting into the while flanks extend outward. In transplantation, alloreactivity arises when recipient T cells recognize foreign MHC molecules directly, often mimicking self-peptide due to structural similarities in the groove, leading to rapid graft rejection. The invariant chain (Ii, or CD74) plays a crucial role in MHC class II maturation by preventing premature peptide binding in the ER and directing the complex to endosomal compartments via its targeting signals. Ii occupies the peptide-binding groove with its CLIP region until degraded by cathepsins, after which HLA-DM catalyzes the exchange for antigenic peptides. Recent structural studies have revealed additional interactions between Ii and MHC class II beyond CLIP, enhancing assembly stability. Non-classical MHC molecules, such as , extend beyond classical pathways, particularly in regulating natural killer () cell activity. presents conserved signal peptides from classical leaders to inhibitory NKG2A/CD94 receptors on cells, inhibiting to maintain self-tolerance. Emerging 2025 research underscores 's role in fine-tuning responses in cancer and transplantation, with structural insights revealing how peptide diversity modulates receptor affinity and functions.

Minor Histocompatibility Antigens

Molecular Basis and Identification

Minor histocompatibility antigens (miHAs) arise from polymorphic proteins encoded by genes outside the (MHC), where allelic variations lead to sequences that differ between individuals and are presented by self-MHC molecules to T cells, potentially triggering alloimmune responses. These , typically 8-11 long, are generated through standard pathways and bind to or II molecules, but their immunogenicity depends on the specific polymorphism creating a T-cell absent in the recipient. A classic example is HA-1, derived from the KIAA0020 gene (also known as the pumilio domain-containing protein), where the immunodominant RTLDKVLEV from the H elicits cytotoxic T-cell responses when , while the L variant (RRLDKVLEV) does not. Similarly, the H-Y antigen originates from the SMCY gene (now termed KDM5D) on the , producing the FIDSYICQV, which is presented by various HLA alleles and recognized by female T cells against male tissues. The first miHA, H-Y, was identified in the 1970s through observations of accelerated skin graft rejection in MHC-matched mice of opposite sexes, establishing the concept of sex-linked histocompatibility disparities beyond MHC control. This discovery highlighted how Y-chromosome-encoded proteins could serve as alloantigens, prompting further studies in both mice and humans. Subsequent identification of miHAs relied on functional assays like to detect interferon-gamma secretion from antigen-specific T cells in response to candidate peptides, often derived from mismatched transplant recipients. Advanced molecular techniques have refined miHA discovery, including applied to HLA-eluted peptides from mismatched grafts or cell lines to directly sequence immunogenic fragments. Genome-wide association studies (GWAS) and whole-genome sequencing, integrated with T-cell reactivity assays, have mapped miHA loci by correlating polymorphisms with immune responses in transplant cohorts. More recently, bioinformatics leveraging projects like the 1000 Genomes has enabled high-throughput prediction of miHAs by scanning for nonsynonymous single-nucleotide polymorphisms that generate predicted MHC-binding peptides with high population mismatch potential. miHAs exhibit diverse specificities, including sex-linked antigens like H-Y, which are absent in females and broadly expressed in tissues, and tissue-restricted variants such as hematopoietic-specific miHAs that preferentially target cells. As of 2024, over 150 miHAs have been identified in humans through these methods, though fewer than 100 are well-characterized with confirmed and clinical relevance, reflecting ongoing challenges in validating expression and epitope dominance.

Contributions to Immune Responses

Minor histocompatibility antigens (miHAs) play a critical role in allorecognition by eliciting T-cell responses that amplify immune reactions beyond (MHC) mismatches. In scenarios where donors and recipients are MHC-identical, miHAs—derived from polymorphic peptides presented by shared MHC molecules—can independently trigger graft rejection, particularly in transplants where even subtle disparities lead to chronic inflammation and tissue damage. These antigens provoke CD8+ and CD4+ T-cell activation, resulting in slower but persistent rejection compared to acute MHC-driven responses, as demonstrated in murine models of and grafting. In graft-versus-host disease (GVHD) following (HSCT), miHA disparities are a primary driver of donor T-cell-mediated attacks on host tissues, especially in HLA-matched settings. Mismatched miHAs, such as those encoded by autosomal genes, stimulate alloreactive T cells to target non-hematopoietic organs like the skin, liver, and , contributing to both acute and forms of GVHD. The H-Y antigen complex, arising from Y-chromosome-encoded proteins, exemplifies this in female-to-male HSCT, where female donor T cells recognize H-Y peptides on male recipient cells, increasing the risk of GVHD with a of 1.55 and associating with higher non-relapse mortality. A 2025 study on molecular disparities of HY antigens further confirms their impact on GVHD and relapse in female-to-male transplants. Beyond transplantation, miHAs influence broader immune responses by modulating T-cell recognition of peptide variants in non-allogeneic contexts. In tumor surveillance, miHA-specific T cells can enhance anti-cancer immunity when tumor cells express disparate alleles, as seen in adoptive therapies where miHA-targeted responses augment graft-versus-leukemia effects without universal GVHD. These effects underscore miHAs' role in fine-tuning adaptive immunity, potentially informing personalized strategies.

Role in Transplantation

Graft Rejection Processes

Graft rejection occurs when the recipient's immune system recognizes the transplanted organ or tissue as foreign, primarily due to mismatches in histocompatibility antigens, leading to immune-mediated destruction of the graft. This process is mediated by both innate and adaptive immune responses, with the major histocompatibility complex (MHC) serving as the primary target for T-cell activation. Rejection is classified into three main types based on timing and underlying mechanisms: hyperacute, acute, and chronic. Hyperacute rejection develops within minutes to hours after transplantation and is triggered by pre-existing antibodies in the recipient that bind to donor antigens, activating complement and causing rapid vascular and . This type is most commonly associated with ABO blood group incompatibility, where anti-A or anti-B isohemagglutinins lead to immediate endothelial damage. Hyperacute rejection is rare in modern practice due to pre-transplant screening but remains irreversible once initiated. Acute rejection typically manifests days to weeks post-transplant, peaking within the first three months, and is predominantly T-cell mediated, involving both cellular and humoral components. It results from the recipient's T cells recognizing mismatched donor MHC molecules, leading to release, , and graft infiltration by lymphocytes. Chronic rejection emerges over months to years, characterized by progressive , vascular occlusion, and , often driven by ongoing low-level immune responses. The key mechanisms of rejection involve allorecognition pathways: direct allorecognition, where recipient T cells directly bind intact donor MHC molecules on graft antigen-presenting cells (APCs), eliciting a robust acute response; and indirect allorecognition, where recipient APCs process and present donor-derived peptides (including MHC peptides) on self-MHC, contributing more to chronic rejection. MHC mismatches, particularly in , -B, and -DR loci, are the principal drivers of acute rejection, as they provoke strong T-cell responses that account for the majority of early graft losses in unmatched transplants. Minor histocompatibility antigens, such as those derived from Y-chromosome genes or tissue-specific proteins, play a lesser but significant role, especially in chronic rejection by sustaining indirect pathway activation and .

Matching Strategies and Outcomes

In solid organ transplantation, particularly kidney transplants, human leukocyte antigen (HLA) matching at the A, B, and DR loci to achieve a 6/6 match is considered ideal, as it significantly enhances graft survival and reduces the incidence of acute rejection episodes. For (HSCT), an 8/8 allelic match at HLA-A, -B, -C, and -DRB1 loci, often extended to include -DQB1 for a 10/10 match, optimizes outcomes by minimizing and improving overall survival rates. Virtual crossmatching, which employs epitope-based algorithms to predict donor-specific compatibility without physical testing, has become a key tool in deceased donor allocation, enabling faster and more precise assessments of immunologic risk. Additional strategies focus on mitigating mismatches when perfect compatibility is unavailable. Permissive mismatches, defined by low-immunogenicity alleles such as those at HLA-DPB1 that share T-cell epitope profiles, allow for safer use of partially mismatched donors in HSCT without substantially increasing rejection risk. For highly sensitized patients with preformed antibodies, desensitization protocols involving , intravenous immunoglobulin, and rituximab reduce antibody levels, facilitating transplantation across HLA barriers while preserving graft function. Improved HLA matching correlates with superior clinical outcomes, including significant reductions in acute rejection rates in kidney transplants and enhanced long-term graft survival across organ types. As of 2025, and Transplantation Network (OPTN) policies, including continuous distribution frameworks, incorporate allele-level resolution in matching algorithms to promote greater in , particularly for underrepresented ethnic groups, by refining allocation to avoid immunogenic disparities. Unrelated donor registries, such as the (NMDP), leverage high-resolution HLA typing to achieve high compatibility rates in donor searches for diverse patient populations—often over 90% for patients of European —expanding transplant opportunities through predictive modeling.

Histocompatibility Testing

Serological Typing Techniques

Serological typing techniques for histocompatibility primarily rely on antibody-based assays to identify (HLA) molecules expressed on the surface of lymphocytes, enabling the detection of serologically defined HLA specificities. These methods emerged as foundational tools in the , with the (CDC) assay, also known as the microlymphocytotoxicity test, developed by Terasaki and McClelland in 1964 as a standardized approach using trays. This technique revolutionized HLA phenotyping by allowing of cell surface antigens through antibody-mediated complement activation and cell . The core process of CDC-based serological typing involves isolating peripheral blood lymphocytes from the individual, which are then incubated in micro-wells with panels of monospecific or broadly reactive anti-HLA antisera targeting known , -B, -C, or - antigens. Following incubation, rabbit complement is added to trigger the if antigen-antibody binding occurs, leading to cell membrane damage and ; viable cells are distinguished from dead ones using vital dyes like or under , with cytotoxicity patterns matched to specific HLA serotypes. For HLA class II typing, B lymphocytes are enriched due to their expression of , , and antigens, as T cells primarily display class I molecules. In crossmatching—a related serological application—recipient is instead incubated with donor lymphocytes to detect pre-existing anti-HLA antibodies that could provoke rejection, following a similar complement readout. Flow cytometry represents an evolution within serological methods, employing fluorescently labeled monoclonal or polyclonal antibodies to bind HLA antigens on lymphocytes, followed by laser-based detection of fluorescence intensity to quantify antigen expression and specificity. This , often integrated as flow cytometric (FCXM), processes cells through a fluid stream for multiparametric analysis, distinguishing T and subsets and providing quantitative data on binding without relying on complement. These serological approaches offer key advantages, including rapid turnaround times—often within hours—making them ideal for urgent pre-transplant in deceased donor scenarios, and their ability to directly assess functional, cell-surface HLA antigens relevant to immune recognition. However, limitations include low , as they define broad serotypes (e.g., HLA-A1) rather than precise alleles, with cross-reactive antibodies leading to up to 25% in assignments, particularly for HLA-B loci. Additionally, the method demands fresh viable cells and skilled interpretation, contributing to its gradual replacement by molecular techniques for high-resolution needs, though it persists in 2025 for quick screening in resource-constrained environments.

Molecular and Advanced Methods

Molecular methods for histocompatibility testing have advanced beyond serological approaches by directly analyzing DNA sequences of (HLA) genes, enabling higher resolution typing at the level. with sequence-specific probes (PCR-SSOP) amplifies HLA loci using locus-specific primers, followed by hybridization to immobilized probes that detect specific sequences, typically resolving alleles to intermediate resolution (e.g., four-digit level) for class I and II genes. Similarly, with sequence-specific primers (PCR-SSP) employs allele-specific primer pairs in multiplex reactions to amplify only matching HLA alleles, visualized via , offering rapid, cost-effective typing suitable for small sample sets with intermediate resolution. These techniques contrast with serological methods by providing genetic rather than phenotypic data, though they may leave some allelic ambiguities in highly polymorphic regions. Next-generation sequencing (NGS) represents a pivotal advancement, allowing comprehensive, full-length sequencing of HLA genes to achieve unambiguous, phase-resolved across multiple loci in a single . NGS workflows typically involve long-range amplification of HLA regions, library preparation, and high-throughput sequencing, followed by bioinformatics to reference sequences, resolving over 95% of potential allelic ambiguities that plague earlier methods. For epitope-level analysis, (MS) techniques, such as liquid chromatography-tandem MS (LC-MS/MS), enable direct identification of HLA-bound peptides from cell surfaces or tissues by immunoaffinity purification of HLA molecules, of associated peptides, and sequencing via MS, revealing immunogenic epitopes without prior knowledge of sequences. This approach has identified thousands of unique HLA-peptide complexes in clinical samples, supporting precise assessment of in transplantation contexts.00073-7/fulltext) Virtual crossmatching integrates these molecular data with computational algorithms to predict donor-recipient compatibility without physical assays, enhancing efficiency in transplant allocation. Tools like HLAMatchmaker use epitope-based matching by parsing HLA alleles into structural epitopes from databases, calculating mismatches at the eplet level to forecast antibody reactivity and immunologic risk, often correlating strongly with physical crossmatch outcomes. Recent advancements, including 2025 proficiency testing programs, emphasize NGS standardization through inter-laboratory comparisons and consensus protocols to ensure reproducibility and minimize reporting discrepancies in clinical HLA typing. The IPD-IMGT/HLA database, updated in 2024, curates over 42,000 allele sequences, facilitating global allele imputation and reference alignment essential for NGS and virtual tools.

Broader Biological Contexts

Evolutionary Significance

The polymorphism observed in histocompatibility genes, particularly the (MHC), has evolved primarily through balancing selection driven by pressures, where allows individuals to recognize and respond to a broader array of s compared to homozygotes. This selective mechanism maintains high allelic diversity at MHC loci, as evidenced by models showing that exposure to multiple s can sustain over 100 alleles per locus under conditions. In vertebrates, MHC diversity traces its origins to approximately 500 million years ago in jawed fish, marking the emergence of the with linked class I and class II genes that provided an ancestral framework for . Comparative studies across species reveal conserved MHC orthologs that underscore this evolutionary continuity; for instance, the H2 complex serves as a well-characterized ortholog to the HLA region, sharing structural and functional similarities in class I and II genes despite genomic rearrangements. In non-mammalian s, such as amphibians, the Xenopus laevis MHC includes nonclassical class Ib genes like those in the XLA family, which exhibit sequence conservation with mammalian counterparts and contribute to immune recognition in early vertebrates. Analogous histocompatibility systems exist in , notably in the colonial ascidian , where a highly polymorphic /histocompatibility (Fu/HC) locus governs vascular or rejection between colonies, mirroring vertebrate allorecognition without adaptive immunity.30337-9) The evolutionary significance of these systems lies in their enhancement of population-level immunity, as MHC polymorphism enables collective to diverse through varied peptide-binding repertoires across individuals. However, this diversity incurs trade-offs, including an increased risk of due to between self and peptides, which constrains MHC expansion and reflects a between and self-tolerance. Recent 2025 reviews on MHC variability highlight parallels with systems, such as and ontogenetic expression patterns, providing models to infer ancient selective pressures on immunity.

Involvement in Autoimmunity and Cancer

Histocompatibility molecules, particularly () class I and II alleles (known as or HLA in humans), significantly influence susceptibility to by modulating peptide presentation to T cells. Specific HLA alleles alter the binding affinity for self-peptides or mimic foreign antigens, leading to dysregulated immune responses against host tissues. For example, the allele is strongly associated with (RA), where it presents arthritogenic peptides more effectively, promoting autoreactive T-cell activation and joint inflammation. Similarly, confers high risk for (AS), with its unique peptide-binding groove facilitating the presentation of self-peptides that trigger chronic inflammation in the spine and sacroiliac joints. These associations highlight how polymorphic variations in MHC molecules can predispose individuals to by enhancing the visibility of self-antigens to the . Mechanisms underlying these links include molecular mimicry and altered peptide binding. Molecular mimicry occurs when microbial peptides structurally resemble self-peptides, allowing cross-reactive T cells to attack host tissues after ; MHC molecules amplify this by presenting the mimicking epitopes to autoreactive T cells. In addition, certain MHC polymorphisms change the peptide-binding repertoire, enabling the stable presentation of autoantigenic peptides that would otherwise be overlooked, thereby breaking . A notable example is in (T1D), where in Mexican populations, the HLA class II DRB10405-DQA10301-DQB1*0302 is linked to increased susceptibility through enhanced presentation of islet autoantigens like insulin or GAD65. In cancer, histocompatibility dynamics contribute to immune evasion and therapeutic targeting. Tumors frequently downregulate expression via mechanisms such as epigenetic silencing by polycomb repressive complex 2 (PRC2), reducing the presentation of tumor antigens to cytotoxic + T cells and allowing from immune . Conversely, neoantigens—mutated peptides unique to cancer cells—are loaded onto MHC molecules for recognition by T cells, forming the basis of personalized immunotherapies like vaccines that boost anti-tumor responses. Minor histocompatibility antigens (miHAs), polymorphic peptides presented by MHC, offer additional avenues for . These antigens can be targeted by chimeric receptor ()-T cells engineered to recognize MHC-bound miHAs, enabling selective killing of tumor cells while sparing healthy tissues in allogeneic settings. Furthermore, alloreactivity— the robust T-cell response against mismatched MHC—has been harnessed in donor infusions to elicit graft-versus-tumor effects, enhancing tumor clearance in hematological malignancies post-transplant. These strategies underscore the dual role of histocompatibility in promoting both tumor evasion and immune-mediated destruction.

References

  1. [1]
    The major histocompatibility complex and its functions - NCBI - NIH
    The function of MHC molecules is to bind peptide fragments derived from pathogens and display them on the cell surface for recognition by the appropriate T ...
  2. [2]
    Genetics, Histocompatibility Antigen - StatPearls - NCBI Bookshelf
    MHC molecules, also known as HLA, are gene-encoded molecules that determine an individual's ability to produce an immune response and are coded by a set of ...
  3. [3]
    The Role of Major Histocompatibility Complex in Organ ...
    Sep 13, 2019 · The MHC plays a dominant role in transplant rejection. On the other hand, the roles of minor histocompatibility antigens are also crucial and ...
  4. [4]
    Types of Stem Cell or Bone Marrow Transplant
    Jul 10, 2025 · Syngeneic stem cell transplant​​ This is a special kind of allo transplant that can only be used if you have an identical sibling (twin or ...
  5. [5]
    Transplant Tolerance, Not Only Clonal Deletion - PMC
    Work of Gorer and Snell, starting in the late 1930s with breeding of congenic strains of mice, identified the Major Histocompatibility Complex (MHC) as the ...
  6. [6]
    4. MHC & Antigen Presentation - Immunopaedia
    The classical HLA antigens encoded in each region include HLA-A, -B, and -C in the MHC-I region, and HLA-DR, -DQ, and -DP in the MHC-II region. The MHC-III ...
  7. [7]
  8. [8]
    Histocompatibility Antigen - an overview | ScienceDirect Topics
    Major histocompatibility antigens vary between individuals; two classes of these antigens are recognized: I MHC antigens and II MHC antigens.
  9. [9]
    H-Y Antigen - an overview | ScienceDirect Topics
    H-Y antigen is defined as a sex-specific minor histocompatibility antigen that may induce rejection of male organs or bone marrow grafts in MHC-identical female ...
  10. [10]
    Clinical impact of H-Y alloimmunity - PMC - NIH
    H-Y antigens are a group of minor histocompatibility antigens encoded on the Y-chromosome with homologous H-X antigens on the X-chromosome.
  11. [11]
    Transplantation Antigen - an overview | ScienceDirect Topics
    Major transplantation antigens, associated with the major histocompatibility complex (MHC), elicit the most rapid rejection responses, while minor antigens ...
  12. [12]
    minor histocompatibility antigens and graft-versus-host disease
    Minor histocompatibility antigens (miHA) are responsible for the occurrence of graft-versus-host disease in the setting of a major histocompatibility complex ...
  13. [13]
    Statistics - IPD-IMGT/HLA Database
    Numbers of HLA Alleles. HLA class I alleles, 29475. HLA class II alleles, 13521. HLA alleles, 42996. Other non-HLA alleles, 1032. Number of confidential alleles ...<|separator|>
  14. [14]
    [PDF] George D. Snell - Biographical Memoirs
    Using the collection, George formulated the “fundamental rules of transplantation”: that tumors could be transplanted freely within an inbred strain, into its ...
  15. [15]
    [PDF] George D. Snell - Nobel Lecture
    One of the first crosses set up, utilizing the three dominant marker genes , Ca, Fu, and W, revealed a linkage between a histocompatibility gene and Fu or fused ...
  16. [16]
    Peter Medawar – Nobel Lecture - NobelPrize.org
    The tolerated CBA graft can be destroyed within a week by injecting into its host, lymphoid cells from A-line mice which have reacted upon and rejected CBA ...Missing: 1940s | Show results with:1940s
  17. [17]
    Jean Dausset (1916–2009) | Nature
    Jul 15, 2009 · In 1952, Dausset made the seminal observation that serum from patients who had received multiple blood transfusions caused marked ...Missing: anti- | Show results with:anti-
  18. [18]
    Jon van Rood: The pioneer and his personal view on the early ...
    The discovery of the HLA antigens as the most important transplantation antigens and their role in the alloimmune response has been crucial for the developments ...
  19. [19]
    Origins of the first HLA specificities - PubMed
    At the 1968 WHO Nomenclature meeting, the naming of the first antigen ultimately gave rise to the designation "HLA" in the human MHC Class I antigen system.Missing: mapping | Show results with:mapping
  20. [20]
    THE HUMAN MAJOR HISTOCOMPATIBILITY COMPLEX AS A ...
    This research was advanced by George Snell who established that graft rejection in mice was due to incompatibility at the level of certain antigens. The murine ...Missing: 1960s | Show results with:1960s
  21. [21]
    History of MHC – 1901 – 1970 - Immunopaedia
    During the first decade of the 20th century, two important advances were made that paved the way to the discovery of the MHC in later years.
  22. [22]
    The Nobel Prize in Physiology or Medicine 1980 - NobelPrize.org
    The Nobel Prize in Physiology or Medicine 1980 was awarded jointly to Baruj Benacerraf, Jean Dausset and George D. Snell for their discoveries.
  23. [23]
    Major histocompatibility complex: Antigen processing and presentation
    The major Histocompatibility complex (MHC) system known as the human leukocyte antigen (HLA) in humans is located on the short arm of chromosome 6 (6p21.3)
  24. [24]
    Chapter 343: The Major Histocompatibility Complex - AccessMedicine
    The human major histocompatibility complex (MHC), commonly called the human leukocyte antigen (HLA) complex, is a 4-megabase (Mb) region on chromosome 6 (6p21.3) ...
  25. [25]
    The HLA genomic loci map: expression, interaction, diversity and ...
    Jan 9, 2009 · The human leukocyte antigen (HLA) super-locus is a genomic region in the chromosomal position 6p21 that encodes the six classical transplantation HLA genes.
  26. [26]
    Genetic diversity, evolution and selection in the ... - BMC Genomics
    Sep 30, 2020 · The MHC loci are known to be among the most polymorphic loci in vertebrate genomes [4]. The generation and long-term maintenance of their ...
  27. [27]
    HLA class II peptide-binding-region analysis reveals funneling of ...
    Highlights · Polymorphism of HLA class II was studied in human population. · Antigen binding pockets (P1, P4, P6, P7, P9) in peptide binding region were selected.
  28. [28]
    IPD-IMGT/HLA Database
    The IPD-IMGT/HLA Database provides a specialist database for sequences of the human major histocompatibility complex (MHC) and includes the official sequences.Allele Query Tool · Sequence Alignment Tool · IPD-MHC · IPD-KIR
  29. [29]
    Advances in the Evolutionary Understanding of MHC Polymorphism
    Oct 25, 2019 · High polymorphism coupled with evidence for positive selection has made MHC genes an attractive model for studying how selection can promote and ...Missing: density | Show results with:density
  30. [30]
    The MHC locus and genetic susceptibility to autoimmune and ...
    Apr 27, 2017 · The class I region includes the three classic human leukocyte antigen (HLA) gene loci: HLA-A, HLA-B, and HLA-C; three non-classic HLA-E, HLA-F, ...
  31. [31]
    "Extended" A1, B8, DR3 haplotype shows remarkable ... - PubMed
    "Extended" A1, B8, DR3 haplotype shows remarkable linkage disequilibrium but is similar to nonextended haplotypes in terms of diabetes risk · Authors.
  32. [32]
    Partial resequencing of 46 distinct HLA haplotypes - ScienceDirect
    This means that the HLA-A1, B8, and DR3 alleles are in strong linkage disequilibrium due to a recent expansion rather than having been derived from distinct ...
  33. [33]
    The ins and outs of MHC class II-mediated antigen ... - PMC
    In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide–MHC class II ...
  34. [34]
    The MHC class I antigen presentation pathway: strategies for viral ...
    This review will discuss recent findings on some of the diverse array of mechanisms employed by human viruses to inhibit the MHC class I pathway in order to ...
  35. [35]
    Major Histocompatibility Complex (MHC) Class I and ... - Frontiers
    Major histocompatibility complex (MHC) class I and class II proteins play a pivotal role in the adaptive branch of the immune system. Both classes of proteins ...<|control11|><|separator|>
  36. [36]
    Endogenous Antigen Presentation of MHC Class II ... - Frontiers
    In the classical MHC class II presentation pathway, exogenous antigens are acquired by antigen-presenting cells and delivered into the endo-lysosomal system ...
  37. [37]
    Peptide length significantly influences in vitro affinity for MHC class II ...
    Since MHC class II molecules have an open-ended binding groove, they do not, in general, restrict the length of bound peptides. The length of peptides occupying ...
  38. [38]
    How an alloreactive T-cell receptor achieves peptide and MHC ...
    Jun 1, 2017 · For example, the lengths and twists of the class I/class II peptide-binding groove α-helices differ, as do the identity of the polymorphic and ...
  39. [39]
    Structural insights into human MHC-II association with invariant chain
    Apr 30, 2024 · Our findings demonstrate that invariant chain not only interacts with MHC-II via class II-associated invariant chain peptide (CLIP) peptide but also engages in ...
  40. [40]
    Review The multifaceted roles of the invariant chain CD74
    The invariant chain (CD74) is well known for its essential role in antigen presentation by mediating assembly and subcellular trafficking of the MHCII complex.
  41. [41]
    HLA‐E: Immune Receptor Functional Mechanisms Revealed by ...
    Jan 3, 2025 · HLA-E is a nonclassical, nonpolymorphic, class Ib HLA molecule. Its primary function is to present a conserved nonamer peptide, termed VL9.
  42. [42]
    HLA-E: Immune Receptor Functional Mechanisms Revealed by ...
    HLA-E is a nonclassical class Ib HLA molecule that presents a peptide to NK cells and T lymphocytes, regulating NK cell function.
  43. [43]
    Minor histocompatibility antigens: time for a paradigm shift?
    Minor histocompatibility antigens (mH-Ags) are polymorphic proteins that are capable of eliciting immune responses between HLA-matched donor and transplant ...
  44. [44]
    Minor Histocompatibility Antigens Are Expressed in ...
    We mapped the expression of four mHAgs (human mHAg 1, pumilio domain-containing protein KIAA0020, B-cell lymphoma 2–related protein A1, and ribosomal protein S4 ...Missing: basis | Show results with:basis
  45. [45]
    The Immunogenicity of a New Human Minor Histocompatibility ...
    To elucidate the basis for differential expression of HA-8 within the human population, we searched protein and DNA sequence databases with the entire KIAA0020 ...
  46. [46]
    HA-1 and the SMCY-derived peptide FIDSYICQV (H-Y ... - PubMed
    HA-1 and the SMCY-derived peptide FIDSYICQV (H-Y) are immunodominant minor histocompatibility antigens after bone marrow transplantation · Authors · Affiliation.Missing: basis KIAA0020
  47. [47]
    Minor Histocompatibility Antigens: from Transplantation Problems to ...
    Aug 5, 2025 · Advanced molecular techniques also permitted the identification of minor histocompatibility antigens (mHags), which play an important role in ...
  48. [48]
    Minor histocompatibility antigens on transfused leukoreduced units ...
    To test this hypothesis, we engineered a mouse model of leukoreduced blood transfusion and MHC-identical–mHA-mismatched bone marrow transplantation. Here, we ...
  49. [49]
    Identification of human minor histocompatibility antigens based on ...
    A robust and efficient method of minor H gene discovery that combines whole genome association scans (WGASs) with cytotoxic T-lymphocyte (CTL) assays, ...
  50. [50]
    Identification of minor histocompatibility antigens based on the 1000 ...
    The 1000 Genomes Project-based identification of minor histocompatibility antigens thus represents a very convenient and robust method for the identification of ...Missing: ELISPOT mass spectrometry
  51. [51]
    Minor Histocompatibility Antigen-Specific T Cells - Frontiers
    Minor Histocompatibility (H) antigens are major histocompatibility complex (MHC)/Human Leukocyte Antigen (HLA)-bound peptides that differ between allogeneic ...Introduction · Minor Histocompatibility... · Examples of HLA Class I Minor...
  52. [52]
    Expanding the repertoire reveals recurrent, cryptic, and ...
    May 2, 2024 · Of all 159 MiHAs, 37 antigens are cryptic, and 11 are new potential targets for immunotherapy due to selective hematopoietic expression.
  53. [53]
    Responses to alloantigens and transplant rejection - NCBI - NIH
    The polymorphic antigens responsible for the rejection of MHC-identical grafts are therefore termed minor histocompatibility antigens or minor H antigens.
  54. [54]
    Minor Histocompatibility Antigen - an overview | ScienceDirect Topics
    Minor histocompatibility antigens are a group of antigens that contribute to the gradual rejection of grafts in transplantation.Histocompatibility · Chaperoning Antigen... · Iv Mhc Class Ii Molecules In...
  55. [55]
    Challenges in GVHD and GVL after hematopoietic stem cell ...
    Aug 21, 2025 · Minor histocompatibility antigens (mHAgs) are potent targets both in GVL and in GVHD. Two recent studies on mHAgs in the GVHD vector deserves ...
  56. [56]
    Minor histocompatibility antigens as targets for immunotherapy ... - NIH
    May 22, 2007 · Minor histocompatibility antigens (mHag) were originally identified as antigens causing graft rejection or graft‐versus‐host disease in ...Missing: influence | Show results with:influence
  57. [57]
    Autosomal Minor Histocompatibility Antigens: How Genetic Variants ...
    These polymorphic peptides or minor histocompatibility antigens (MiHA) are produced by genetic differences between patient and donor.
  58. [58]
    The Major Histocompatibility Complex in Transplantation - PMC - NIH
    In organ transplantation, the adaptive immunity is considered the main response exerted to the transplanted tissue, since the principal target of the immune ...
  59. [59]
    Acute Transplantation Rejection - StatPearls - NCBI Bookshelf - NIH
    Acute transplantation rejection is an immune response against the donor graft that causes tissue impairment and potential failure.[1] T-cell and antibody- ...
  60. [60]
    Immunology of Transplant Rejection - Medscape Reference
    Jul 11, 2023 · In hyperacute rejection, the transplanted tissue is rejected within minutes to hours because vascularization is rapidly destroyed. Hyperacute ...
  61. [61]
    HLA Mismatching Strategies for Solid Organ Transplantation - PMC
    HLA matching provides numerous benefits in organ transplantation including better graft function, fewer rejection episodes, longer graft survival, ...
  62. [62]
    Optimal HLA Matching in Haematopoietic Cell Transplantation - PMC
    [6] These data suggest that outcome after HCT from “8/8” matched donors at HLA-A, B, C and DRB1 is associated with similarly good outcome as 10/10 HCT, and ...
  63. [63]
    Principles of Virtual Crossmatch Testing for Kidney Transplantation
    Virtual crossmatch (VXM) test assesses immunologic compatibility between recipient and potential donor by analyzing the results of 2 independently done physical ...
  64. [64]
    HLA-C expression levels define permissible mismatches in ... - NIH
    The similarity of sequence features between allele mismatches may contribute to their lower immunogenicity. A second model of HLA-C alloreactivity entails ...
  65. [65]
    Current Approaches to Desensitization in Solid Organ Transplantation
    Use of Intravenous Immune Globulin and Rituximab for Desensitization of Highly HLA-sensitized Patients Awaiting Kidney Transplantation. Transplantation (2010) ...
  66. [66]
    Significance of HLA-DQ in kidney transplantation: time to reevaluate ...
    Jul 7, 2021 · We address the implications of reduced HLA matching policies in kidney allocation. We highlight the observed diminished outcome data, the significant financial ...
  67. [67]
    Continuous Distribution of Kidneys, Winter 2025 - OPTN - HRSA
    The inclusion of high-resolution HLA typing in the database is a first step in improving matching and avoiding delays in organ allocation due to the presence of ...
  68. [68]
    HLA Match Likelihoods for Hematopoietic Stem-Cell Grafts in the ...
    Jul 24, 2014 · The success of unrelated-donor HSCT is greatly influenced by the degree of HLA matching between the donor or cord-blood unit and the recipient.
  69. [69]
    Complement‐Dependent Cytotoxicity (CDC) to Detect Anti‐HLA ...
    The method to detect anti‐HLA antibodies has historically been the complement‐dependent cytotoxicity (CDC) assay, described by Terasaki and McClelland in 1964 3 ...
  70. [70]
    A walk through the development of human leukocyte antigen typing
    In this review, we explore the developmental history and future prospects of HLA typing technology, which promises to further benefit the field of ...
  71. [71]
  72. [72]
    Complement-Dependent Cytotoxicity (CDC) to Detect Anti-HLA ...
    Aug 6, 2025 · The criterion (gold) standard to detect anti-human leukocyte antigen (HLA) antibodies is the complement-dependent cytotoxicity (CDC) assay.
  73. [73]
    Development of PCR-SSOP for the Identification of HLA-A*02 ...
    A PCR-SSOP typing method, involving a single PCR amplification in conjunction with 19 digoxigenin labelled oligonucleotide probes, has been developed for the ...Missing: technique | Show results with:technique
  74. [74]
    HLA Typing by Sequence-Specific Primers
    PCR-SSP is a commonly used technique in HLA typing in which multiple pairs of cis-located allele-specific primers are used to determine the alleles present in ...
  75. [75]
    Advancements in HLA Typing Techniques and Their Impact on ...
    Mar 5, 2024 · SSP typing is generally considered to provide intermediate resolution. This technique is efficient for typing a small number of samples quickly ...
  76. [76]
    HLA Sequencing | HLA typing with NGS - Illumina
    HLA typing by next-generation sequencing (NGS) generates unambiguous, phase-resolved HLA typing results using a single assay, system, and analysis program.
  77. [77]
    OptiType: precision HLA typing from next-generation sequencing data
    OptiType is a novel HLA genotyping algorithm using integer linear programming for accurate HLA typing from NGS data, achieving 97% accuracy.
  78. [78]
    Mass Spectrometry Profiling of HLA-Associated Peptidomes in Mono ...
    Feb 21, 2017 · Identification of human leukocyte antigen (HLA)-bound peptides by liquid chromatography-tandem mass spectrometry (LC-MS/MS) is poised to ...
  79. [79]
    Comparison of Physical Crossmatch and Virtual ... - PubMed Central
    Feb 3, 2019 · The results of human leukocyte antigen (HLA) epitope analysis using virtual crossmatch (VXM) with HLAMatchmaker show serological donor ...
  80. [80]
    Editorial: Proficiency testing in histocompatibility and immunogenetics
    Jun 4, 2025 · This article is part of the Research Topic Proficiency testing in Histocompatibility and Immunogenetics: current status and future perspectives
  81. [81]
    How pathogens drive genetic diversity: MHC, mechanisms and ...
    The heterozygote advantage hypothesis proposes that individuals heterozygous at MHC loci are able to respond to a greater range of pathogen peptides than ...
  82. [82]
    Heterozygote advantage can explain the extraordinary diversity of ...
    Nov 26, 2024 · We here present an eco-evolutionary model showing that evolution can result in the emergence and maintenance of more than 100 alleles under HA.
  83. [83]
    Origin and evolution of the adaptive immune system - NIH
    ... 500 million years ago in jawed fish. This intricate defence system consists of many molecules, mechanisms and tissues that are not present in jawless ...
  84. [84]
    An Ancestral Major Histocompatibility Complex Organization in ...
    Dec 6, 2023 · Our results on the elasmobranch MHC strongly suggest that all MHC genes were originally linked together and had a common origin, in the jawed ...Results And Discussion · A Complement C4 Gene (c4a)... · A Proto-Mhc/agr Emerged At...
  85. [85]
    Comparative genomics of the human, macaque and mouse major ...
    Jul 10, 2016 · We provide a short review of the genomic similarities and differences among the human, macaque and mouse MHC class I and class II regions.
  86. [86]
    Remarkable Conservation of Distinct Nonclassical MHC Class I ...
    We have conducted an extensive genomic, molecular, and phylogenetic characterization of class Ib genes in two Xenopodinae amphibian species of different genera ...Missing: H2 XLA<|separator|>
  87. [87]
    Histocompatibility in Botryllus schlosseri and the origins of adaptive ...
    May 10, 2025 · The basal chordate, Botryllus schlosseri, undergoes a natural transplantation reaction that is controlled by a single, highly polymorphic locus called the fuhc.
  88. [88]
    The importance of immune gene variability (MHC) in evolutionary ...
    Oct 20, 2005 · MHC variability is believed to be maintained by pathogen-driven selection, mediated either through heterozygote advantage or frequency-dependent ...Selection Mechanisms · Importance Of Mhc... · Relevance Of Mhc...
  89. [89]
    The Amphibian Major Histocompatibility Complex—A Review and ...
    Jan 7, 2025 · The MHC region comprises three subregions: MHC class I, MHC class II and MHC class III, and encodes about 200 genes in mammals and amphibians ( ...<|separator|>
  90. [90]
    Association of MHC and rheumatoid arthritis: HLA-DR4 ... - PMC - NIH
    Inherited susceptibility to rheumatoid arthritis (RA) is associated with the DRB1 genes encoding the human leukocyte antigen (HLA)-DR4 and HLA-DR1 molecules.
  91. [91]
    Fifty years after the discovery of the association of HLA B27 with ...
    The association of HLA B27 alleles with ankylosing spondylitis (AS), a highly heritable disease, which is part of the spectrum of axial spondyloarthritis (axSpA) ...
  92. [92]
    Molecular Mimicry as a Mechanism of Autoimmune Disease - PMC
    Inflammation induced by exposure to a foreign antigen can lead to autoimmune diseases from cross-reactive epitopes (molecular mimicry). These epitopes are ...
  93. [93]
    MHC Class II Polymorphisms, Autoreactive T-Cells, and Autoimmunity
    Antigenic peptides are embedded in the peptide-binding groove of the MHC ... peptide-binding pocket predispose to autoimmune thyroiditis in humans and in mice.
  94. [94]
    HLA and autoimmune diseases: Type 1 diabetes (T1D) as an example
    The "Diabetogenic haplotypes" in Mexicans were DRB1*0301-DQA1*0501-DQB1*0201 (OR = 21.4); DRB1*0405-DQA1-*0301-DQB1*0302 (OR = 44.5) and the same DQA1/DQB1 with ...
  95. [95]
    PRC2-mediated MHC-I silencing drives immune evasion - Nature
    Oct 15, 2019 · An evolutionarily conserved function of Polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer.
  96. [96]
  97. [97]
    TCR-Like CAR-T Cells Targeting MHC-Bound Minor ...
    Feb 28, 2020 · This mini review focuses on the potential for and limitations of TCR-like CAR-T cells in targeting human leukocyte antigen–bound peptide antigens.
  98. [98]
    Immunotherapy of cancer with alloreactive lymphocytes - PubMed
    Immunotherapy of cancer with alloreactive lymphocytes is the mainstay of treatment, especially in haematological malignant disease.