Fact-checked by Grok 2 weeks ago

MHC class I

Major histocompatibility complex (MHC) class I molecules are transmembrane glycoproteins expressed on the surface of nearly all nucleated cells in vertebrates, functioning to display short peptides derived from intracellular proteins to cytotoxic CD8+ T cells as part of the adaptive immune response. These molecules enable the immune system to distinguish healthy cells from those infected by viruses, transformed by cancer, or otherwise compromised, thereby triggering targeted destruction of aberrant cells while inhibiting natural killer (NK) cells through recognition of self-peptides. Structurally, MHC class I consists of a polymorphic α heavy chain non-covalently associated with the invariant β2-microglobulin (β2m) light chain; the α chain features three extracellular domains (α1, α2, and α3), where the α1 and α2 domains form a peptide-binding groove that accommodates peptides typically 8–10 amino acids in length, anchored by specific residues in polymorphic pockets. The biosynthesis and antigen presentation pathway of MHC class I molecules occur primarily in the (ER), where newly synthesized heavy chains associate with chaperones like and before β2m binding; peptides generated in the by the are transported into the ER via the transporter associated with antigen processing (), trimmed by endoplasmic reticulum aminopeptidases (ERAPs), and loaded onto MHC class I with assistance from the peptide-loading complex, including tapasin. This process ensures stable peptide-MHC complexes are transported to the cell surface via the Golgi apparatus for immune surveillance. Genetically, the genes encoding human MHC class I (known as human leukocyte antigens or , , and ) are located in the MHC region on the short arm of , exhibiting extreme polymorphism with over 29,000 alleles identified (as of September 2025), which enhances population-level immune diversity by allowing presentation of a broad repertoire of peptides; inheritance is codominant and follows Mendelian patterns. Evolutionarily, MHC class I molecules emerged around 500 million years ago, with their polymorphism maintained through balancing selection, including pathogen-driven pressures and preferences. In addition to classical MHC class I, non-classical variants like , HLA-F, and play specialized roles, such as modulating NK cell activity or contributing to during pregnancy. While ubiquitous in expression, levels of MHC class I can be downregulated by certain viruses or tumors to evade detection, underscoring their central role in immune homeostasis.

Structure

Molecular Components

The major histocompatibility complex (MHC) class I molecule is composed of a polymorphic heavy chain, also known as the alpha chain (α chain), which is a with an approximate molecular weight of 45 kDa. This heavy chain consists of three extracellular domains—α1, α2, and α3—a transmembrane region that anchors it to the , and a short cytoplasmic tail. In humans, the heavy chain is encoded by one of the highly polymorphic HLA-A, HLA-B, or , located within the MHC region on chromosome 6. Non-covalently associated with the heavy chain is β2-microglobulin (β2m), a non-polymorphic light chain with a molecular weight of approximately 12 that is essential for the of the MHC class I complex. The β2m subunit is encoded by the separate B2M gene on and shares structural similarity with the immunoglobulin domain, particularly resembling the α3 domain of the heavy chain. The heavy chain undergoes post-translational modifications, including N-linked glycosylation primarily at a conserved residue (Asn86) in the α1 domain, which contributes to , , and trafficking within the . This glycosylation site is present across different HLA alleles, though the extent of glycan processing can vary.

Architecture and Domains

The (MHC) class I molecule forms a heterodimeric complex composed of a polymorphic heavy chain, also known as the α chain, and the invariant light chain β₂-microglobulin (β₂m). The heavy chain consists of three extracellular domains—α₁, α₂, and α₃—linked to a transmembrane and a short cytoplasmic tail, while β₂m associates non-covalently with the extracellular portion. The α₁ and α₂ domains, each comprising approximately 90 , fold into a β-sheet platform topped by α- that create a cleft for binding, whereas the membrane-proximal α₃ domain adopts an immunoglobulin-like fold essential for interaction with the co-receptor on cytotoxic T cells. The structural integrity of this platform relies on intimate interactions between the domains and β₂m. β₂m binds to the underside of the α₁-α₂ platform, stabilizing the heavy chain through extensive bonding networks involving conserved residues, such as those in the β-strands of β₂m with complementary regions in α₁ and α₂. Additionally, an intra-domain bridge (Cys101-Cys164) within the α₂ , along with inter-domain bonds, maintains the helical architecture of the -binding cleft, preventing unfolding in the absence of . These interactions ensure the molecule's stability on the surface, with β₂m playing a critical role in folding and assembly. The three-dimensional structure of MHC class I was first elucidated in 1987 through of HLA-A2 at 3.5 Å resolution by Bjorkman et al., revealing a closed-ended groove formed by the α₁-α₂ helices, ideally suited for accommodating of 8-10 residues in an extended conformation. This landmark study demonstrated how the platform elevates the for T , with the α₃ positioned below to facilitate co-receptor engagement. Subsequent higher-resolution structures have refined these insights, confirming the conserved topology across (HLA) and mouse H-2 alleles. MHC class I molecules exhibit conformational flexibility, adopting open or closed states influenced by peptide occupancy. In the peptide-free or low-affinity state, the α-helices of the cleft partially separate, widening the groove to facilitate peptide entry, as observed in crystal structures of empty HLA-A*02:01. High-affinity peptide binding induces a closed conformation, where the helices converge, locking the peptide via hydrogen bonds and van der Waals interactions at the termini, thereby enhancing surface stability and immune surveillance. These dynamic transitions underscore the molecule's role in antigen presentation efficiency.

Peptide Binding Groove

The peptide-binding groove of MHC class I molecules is a key structural feature located at the interface of the α1 and α2 domains, formed by two parallel α-helices that flank a floor composed of an eight-stranded antiparallel β-sheet. This architecture creates a cleft approximately 25 long and 12 wide, designed to bind antigenic derived from intracellular proteins. The groove's closed ends, enforced by conserved residues, restrict peptide length to typically 8-10 , ensuring stable presentation on the surface. Within the groove, peptides are anchored primarily through interactions with specific pockets that accommodate side chains at defined positions. The A pocket at the N-terminal end binds the peptide's amino group via conserved hydrogen bonds, while the F pocket at the C-terminal end interacts with the carboxyl group and a hydrophobic residue. For example, in HLA-A*02:01, the F pocket favors a C-terminal or , contributing to its preference for nonamer peptides with motifs like L/M at position 2 and V/L at the C-terminus. Additional pockets (B through E) accommodate secondary anchors and variable residues, allowing peptide bulging for lengths up to 11-12 without disrupting overall binding. Polymorphisms in MHC class I alleles primarily cluster in these pockets, altering specificity and the of presented . Variations in pocket depth and residue composition can enhance or restrict anchor preferences; for instance, HLA-B27's deep B , lined by at position 77 and other residues, selectively binds peptides with at position 2, which is linked to its role in spondyloarthropathies. Such allelic differences ensure diverse immune across populations by modulating peptide selectivity without compromising groove stability. Peptides adopt an extended, polyproline II-like conformation within the groove, aligning parallel to the β-sheet floor in a manner resembling an additional antiparallel strand. This is stabilized by a network of invariant hydrogen bonds from conserved residues—such as Tyr7 in the β-sheet, Tyr59 in the α1 , Tyr159 in the α2 , and Tyr171—to the peptide's main-chain atoms, particularly at positions 1, 2, the , and penultimate residue. For peptides longer than nine residues, central bulges allow accommodation while maintaining anchor contacts, preserving the overall structural integrity essential for T recognition.

Biosynthesis and Assembly

Intracellular Synthesis

MHC class I genes exhibit constitutive expression in nearly all nucleated cells, driven by conserved promoter elements including enhancer A, which binds , and the interferon-stimulated response element (ISRE), which interacts with interferon regulatory factor (IRF) family members. This basal transcription is further modulated by the SXY module, forming an enhanceosome with transcription factors such as RFX, CREB/ATF, and NF-Y to maintain steady-state levels essential for immune surveillance. Inducible expression is primarily triggered by -gamma (IFN-γ), which activates the JAK/STAT pathway, leading to phosphorylation and subsequent induction of IRF1; these factors bind to the ISRE and gamma-activated site (GAS) elements, significantly upregulating MHC class I transcription during immune responses. The heavy chain and β2-microglobulin (β2m), a non-covalently associated light chain essential for MHC class I stability, are both synthesized on free ribosomes in the cytosol. The heavy chain, encoded by HLA-A, -B, or -C genes, features an N-terminal signal peptide that directs its co-translational translocation into the endoplasmic reticulum (ER) lumen via the Sec61 translocon, where the signal peptide is cleaved to initiate membrane integration. β2m also features an N-terminal signal peptide that directs its co-translational translocation into the ER lumen via the Sec61 translocon, where the signal peptide is cleaved, allowing it to associate with the heavy chain during subsequent assembly steps. Upon entry into the ER, the nascent heavy chain undergoes initial folding, beginning with binding to the lectin chaperone , which recognizes the monoglucosylated N-linked on the α3 domain to facilitate and prevent aggregation. Disulfide bond formation in the α1 and α2 domains, critical for the peptide-binding groove , is catalyzed by the oxidoreductases ERp57 and (PDI), often in complex with or . Unfolded or misfolded nascent heavy chains are subject to rapid ER-associated degradation (ERAD), with a of approximately 30-60 minutes, ensuring efficient turnover and preventing accumulation of defective molecules.

Translocation to ER

Cytosolic proteins are primarily degraded by the 26S proteasome into short peptides, typically ranging from 8 to 11 in length, which serve as precursors for MHC class I . This process generates a diverse pool of peptides from ubiquitinated proteins, with the proteasome's catalytic core, the particle, cleaving internal bonds to produce these fragments. Under inflammatory conditions, interferon-gamma (IFN-γ) induces the formation of immunoproteasomes by incorporating specialized subunits such as LMP2 (β1i) and LMP7 (β5i), which alter the cleavage specificity to favor the production of peptides suitable for MHC class I binding. The transporter associated with antigen processing (TAP), a member of the ATP-binding cassette (ABC) transporter family, facilitates the translocation of these cytosolic peptides into the endoplasmic reticulum (ER) lumen. TAP forms a heterodimer consisting of TAP1 and TAP2 subunits embedded in the ER membrane, each contributing six transmembrane domains and a nucleotide-binding domain for ATP hydrolysis. Peptide binding occurs at a specific site in the ER-facing transmembrane domains, with TAP exhibiting selectivity for peptides bearing hydrophobic or basic residues at their C-terminus, ensuring compatibility with MHC class I groove preferences. ATP hydrolysis powers the conformational changes necessary for peptide transport across the membrane, with each cycle driven by the sequential binding and hydrolysis of two ATP molecules per subunit. Upon , peptides are translocated into the at a rate of approximately 100 peptides per minute per complex, enabling efficient supply for MHC class I loading. The affinity of peptides for can be influenced by N-terminal trimming in the by endoplasmic reticulum 1 (ERAP1), which processes longer precursors (often 9-16 residues) transported by into optimal 8-10 residue lengths, thereby modulating the available peptide repertoire. Viral pathogens have evolved mechanisms to evade this process; for instance, the human cytomegalovirus (HCMV) glycoprotein US6 binds to the ER-luminal side of , inhibiting ATP and to block peptide translocation and reduce MHC class I surface expression.

Peptide Loading and Editing

Peptide loading onto MHC class I molecules occurs in the (ER) following the translocation of cytosolic via the transporter associated with antigen processing (TAP). This process ensures that only high-affinity , typically 8-10 long, are selected to stabilize the MHC class I complex for surface . The (PLC), a multi-protein , orchestrates this selection by bridging TAP to newly synthesized MHC class I heavy chains associated with β2-microglobulin (β2m). The PLC comprises tapasin, ERp57 (a ), and , which collectively chaperone MHC class I folding and . Tapasin, an ER-resident glycoprotein, recruits MHC class I to , facilitating access to the pool and promoting iterative to favor high-affinity ligands. ERp57 and assist in maintaining proper bonds and lectin-like to monoglucosylated MHC class I, respectively, enhancing the stability of the loading platform. Concurrently, endoplasmic reticulum aminopeptidases (ERAP1 and ERAP2) trim the N-termini of imported to optimize fit within the MHC class I groove, independently editing length and sequence for better anchor residue compatibility. During loading, empty MHC class I molecules are conformationally unstable and adopt an "open" form, exposing the peptide-binding groove for exchange, while high-affinity peptide binding induces a "closed" conformation that locks the complex. Tapasin preferentially binds peptide-deficient MHC class I, catalyzing the removal of low-affinity peptides and replacement with superior ones through this conformational plasticity. This editing ensures immunodominance of peptides with optimal kinetics. Quality control mechanisms retain unloaded or suboptimally loaded MHC class I in the via retention signals and chaperone interactions, preventing premature export. Approximately 50% of MHC class I molecules are degraded in the if they remain peptide-free, primarily through ER-associated degradation (ERAD) pathways involving retrotranslocation to the and proteasomal . Only peptide-loaded complexes achieve sufficient stability for release from the and progression to the Golgi.

Antigen Presentation Mechanism

Surface Expression and Stability

Following successful peptide loading within the in the , MHC class I molecules dissociate from the PLC and are exported from the ER via COPII-coated vesicles. This release typically occurs upon binding of high-affinity peptides, ensuring only stable complexes proceed in the secretory pathway. The loaded MHC class I complexes then traverse the Golgi apparatus through the conventional secretory route, where the N-linked glycan on the heavy chain (at Asn86) undergoes maturation, including initial trimming of residues by mannosidase I in the cis-Golgi compartment. This processing step refines the high-mannose glycan acquired in the ER into complex forms as the molecules advance through the medial- and trans-Golgi networks. From the trans-Golgi network, peptide-loaded MHC class I molecules are packaged into secretory vesicles and transported to the plasma membrane for insertion. On the surface, these complexes achieve a typical density of approximately 10^5 to 10^6 molecules per , varying by and physiological conditions. This level of expression supports efficient surveillance by cytotoxic T cells and natural killer cells. Surface residency is not permanent; MHC class I molecules undergo constitutive , primarily via clathrin-independent mechanisms involving Arf6, with rates influenced by their conformational stability. The stability of surface MHC class I is predominantly governed by the affinity of the bound , which dictates the complex's , ranging from hours for low-affinity interactions to days for high-affinity ones. Empty or peptide-receptive MHC class I molecules, lacking stable ligands, exhibit rapid internalization through and are prone to or retrieval, preventing unproductive surface . Following , internalized complexes enter early endosomes for sorting: stable peptide-loaded forms are often directed to endosomes (marked by Rab11a and Rab22a) for return to the plasma membrane, while unstable ones may proceed to late endosomes or lysosomes for via the multivesicular body pathway. In some cases, peptide-receptive MHC class I can be retrotranslocated from post- compartments back to the , mediated by chaperones like TAPBPR, allowing for peptide re-editing and potential reloading.

Interaction with T Cell Receptor

The interaction between MHC class I molecules presenting antigenic peptides (pMHC) and the (TCR) on CD8+ T cells is a of adaptive immune . The structural basis of this interaction was first elucidated through of the human TCR A6 bound to HLA-A2 presenting the HTLV-1 peptide, revealing a diagonal mode where the TCR sits atop the pMHC complex at an approximately 45-degree angle relative to the peptide-binding groove. In this orientation, the complementarity-determining regions (CDRs) of the TCR α and β chains primarily contact the α1 and α2 helices of the MHC class I heavy chain, with CDR3 loops focusing on the exposed residues to confer specificity. Additionally, the TCR α chain interacts with the peptide, while the β chain engages the MHC helices more extensively, enabling between self and foreign peptides. The binding interface extends beyond the TCR-pMHC contacts to include the co-receptor, which binds to the α3 domain of the MHC class I molecule, stabilizing the overall complex and facilitating . The TCR α/β heterodimer makes direct contacts with the peptide-MHC via its variable , with the α3 domain serving as the primary docking site for the α/α or α/β homodimers or heterodimers expressed on cytotoxic T cells. This co-receptor engagement enhances the of the interaction, as binding occurs independently of the TCR-pMHC contact but with distinct kinetics, exhibiting a low (K_d ≈ 0.2 at 37°C) that supports rapid association and dissociation. The combined TCR-pMHC and -α3 interactions position the TCR for precise surveillance on the cell surface. The of TCR for pMHC complexes typically ranges from 1 to 100 μM, reflecting a balance between specificity and sensitivity that allows detection of rare antigens. This moderate arises from the structural complementarity at the , where germline-encoded polymorphisms in MHC class I alleles influence and can lead to alloreactivity, as seen in where donor MHC variants are recognized as foreign by host T cells. Such alloreactivity stems from the mimicry of self-pMHC by allogeneic MHC loaded with self-s, driven by sequence differences in the α1 and α2 domains. The specificity is further tuned by the TCR's ability to cross-react with similar s, enabling broad immune coverage without excessive autoreactivity. Upon pMHC engagement, TCR clustering on the T cell surface initiates signaling by recruiting and activating the , which is associated with the co-receptor. This clustering amplifies weak individual interactions into a multivalent array, promoting Lck-mediated phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) on the CD3 ζ chains. The process aligns with the kinetic proofreading model, wherein multiple enzymatic steps impose a time delay, ensuring that only sustained TCR-pMHC engagements (lasting seconds to minutes) lead to productive signaling, while brief encounters dissociate without activation. This mechanism enhances discrimination between and peptides, underpinning the specificity of T cell responses.

Peptide Removal and Recycling

Peptide dissociation from MHC class I molecules occurs at a pH-dependent rate, with stable complexes exhibiting half-lives typically ranging from 1 to 10 hours at neutral on the surface. In acidic endosomal environments ( ≈5.0-6.0), dissociation accelerates significantly—up to 100-fold—due to stabilization of a -empty intermediate, facilitating rapid off-rates without complete heavy chain-β2-microglobulin . Surface , which correlates with , influences removal rates, as more stable -MHC complexes resist and turnover. Internalized peptide-MHC class I complexes are trafficked via clathrin-independent to early sorting endosomes, where they may undergo sorting decisions. A portion is directed to late endosomes and lysosomes for proteolytic degradation, while another fraction recycles back to the plasma membrane through Rab11-positive recycling endosomes, allowing reuse in . This recycling pathway, involving like Arf6, enables dynamic turnover and contributes to the maintenance of surface peptide diversity. During recycling, dissociated peptides or newly generated fragments can undergo brief re-trimming by endosomal proteases, such as insulin-regulated (IRAP), which removes N-terminal residues to optimize fit within the MHC class I groove. This process helps sustain a diverse and immunogenic repertoire on recycled molecules, particularly in antigen-presenting cells. Unlike , which relies on for catalyzed in endosomes, MHC class I lacks equivalent DM-like inhibitors or chaperones, making its endosomal primarily pH- and protease-driven.

Immune Functions

Cytotoxic T Cell Activation

, also known as , are activated when their (TCR) recognizes antigenic peptides presented by MHC class I molecules on the surface of infected or abnormal cells. This TCR engagement provides signal 1 for T cell activation, but full activation requires co-stimulation through the receptor on the T cell interacting with B7-1 () or B7-2 () ligands on antigen-presenting cells, delivering signal 2. Together, these signals trigger intracellular signaling cascades, including activation of and NFAT pathways, leading to the production of interleukin-2 (IL-2) and expression of the alpha chain. IL-2 then drives clonal proliferation and differentiation of naive into effector cytotoxic T lymphocytes (CTLs). Upon differentiation, CTLs acquire effector functions to eliminate target cells. The primary mechanism involves the release of cytotoxic granules containing perforin and granzymes through at the . Perforin forms pores in the target , allowing granzymes to enter and activate , leading to . Additionally, CTLs express (FasL), which binds Fas on target cells to induce death receptor-mediated via the extrinsic pathway. These mechanisms ensure precise of antigen-bearing cells while minimizing bystander damage. Effective CTL activation requires a threshold of TCR-pMHC interactions to initiate signaling above the threshold. This serial engagement model allows a single to activate multiple T cells efficiently. Over the course of an , + T cells undergo avidity maturation, where their functional to peptide-MHC complexes increases up to 50-fold without necessarily selecting for higher-affinity TCRs, enhancing responsiveness to low-antigen levels. Activated CD8+ T cells differentiate into memory subsets that provide long-term immunity. Central memory CD8+ T cells reside in lymphoid organs and exhibit high proliferative potential upon re-encountering , while effector memory CD8+ T cells patrol peripheral tissues for rapid effector responses. These memory cells persist for years post-infection through homeostatic and IL-7/IL-15 signaling, enabling faster and more robust secondary responses.

NK Cell Regulation

MHC class I molecules play a in regulating natural killer () cell activity through the "missing self" hypothesis, which posits that cells detect and eliminate cells lacking sufficient self MHC class I expression, thereby removing inhibitory signals that normally prevent -mediated . This concept emerged from observations that cells reject tumor variants deficient in H-2 (the equivalent of MHC class I) but spare those expressing normal levels, suggesting an alternative immune surveillance mechanism beyond adaptive responses. In healthy cells, surface MHC class I molecules engage inhibitory receptors on cells, setting a threshold for activation; their absence, as seen in virally infected or transformed cells, disarms this inhibition and licenses cell killing. Central to this regulation are interactions between killer cell immunoglobulin-like receptors (KIRs) on cells and specific epitopes on , the primary MHC class I ligand for human KIRs. HLA-C allotypes are grouped into C1 (characterized by at position 80) and C2 (lysine at position 80), with inhibitory KIR2DL2 and KIR2DL3 preferentially binding C1 epitopes, while KIR2DL1 binds C2 epitopes. Upon ligand engagement, these inhibitory KIRs transmit signals via immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in their cytoplasmic tails, which recruit tyrosine phosphatases such as SHP-1, dephosphorylating activation signaling pathways and dampening cell responses like and production. This specificity ensures that cells from individuals expressing particular HLA-C groups are tuned to recognize deviations in self MHC expression. Balancing these inhibitory interactions, certain activating ligands on MHC class I molecules or non-classical variants can counteract inhibition under specific conditions. For instance, rare HLA alleles may engage activating KIRs like KIR2DS1, which shares structural similarity with KIR2DL1 but signals through ITAM-containing adaptors to promote activation. Non-classical MHC molecules, such as , bind heterodimeric CD94/NKG2 receptors on cells; while the NKG2A isoform delivers inhibition similar to KIRs, the NKG2C isoform can activate cells, particularly in contexts like infection where presents viral peptides. These activating interactions provide a rheostat-like balance, preventing over-inhibition while maintaining tolerance to healthy self cells. NK cell responsiveness is further calibrated during development through a process known as education or licensing, where interactions with self MHC class I ligands set the activation threshold for mature NK cells. NK cells expressing inhibitory receptors that bind self HLA molecules (e.g., KIR2DL1 in C2-positive individuals) become "educated" and hyporesponsive to targets lacking those ligands, ensuring functional competence only against truly abnormal cells. This tuning mechanism, observed in both human and mouse models, prevents autoimmunity by rendering uneducated NK cells anergic, while allowing educated ones to mount calibrated responses proportional to the strength of self MHC interactions during ontogeny.

Immune Surveillance Role

MHC class I molecules play a central role in immune surveillance by presenting endogenous peptides derived from intracellular proteins on the cell surface of nearly all nucleated cells, allowing cytotoxic + T cells to monitor for signs of cellular abnormality. This presentation enables the to discriminate between healthy self cells and those compromised by intracellular pathogens or oncogenic mutations, as altered protein degradation products—such as proteins or neoantigens from mutated genes—are loaded onto MHC class I for . In infections, for instance, peptides from pathogen-derived proteins are processed by the and displayed, signaling infection to trigger targeted cell lysis. Similarly, in cancer, MHC class I showcases tumor-specific peptides arising from genetic alterations, facilitating immune detection and elimination of malignant cells. Quantitative analyses of the MHC class I peptidome in infected cells reveal that peptides occupy a small but immunologically significant proportion of surface MHC class I molecules, sufficient to elicit robust T cell responses. This low abundance underscores the sensitivity of the mechanism, where even minor shifts in peptide can alert patrolling + T cells to initiate protective . In tumor contexts, analogous low-level presentation of neoantigens supports ongoing immune oversight, preventing unchecked proliferation. A key extension of this is , primarily by dendritic cells, which internalize exogenous antigens—such as those from apoptotic infected cells or tumor debris—and process them for loading onto MHC class I via specialized pathways like endosome-to-cytosol translocation or vacuolar degradation. This mechanism bridges extracellular threats to + T cell priming, enabling immune responses against viruses that evade direct of antigen-presenting cells or against non-replicating tumor antigens, thereby broadening systemic . To prevent while maintaining vigilance, MHC class I contributes to central through AIRE ()-driven expression of tissue-specific self-antigens in medullary thymic epithelial cells, where these antigens are presented to developing thymocytes for negative selection of autoreactive + T cells. This process ensures that only T cells tolerant to self-peptides mature, striking a balance that supports effective discrimination of non-self threats without aberrant self-attack.

Specialized Physiological Roles

Maternal-Fetal Tolerance in Reproduction

In the context of maternal-fetal tolerance, extravillous trophoblast cells at the placental interface express the non-classical MHC class I molecule , while avoiding expression of classical , , and molecules, thereby minimizing recognition and attack by maternal cytotoxic T lymphocytes (CTLs). This selective expression pattern is crucial for shielding the semi-allogeneic from maternal alloreactive immune responses, as HLA-G's restricted presentation of peptides differs from the diverse repertoire of classical MHC class I, reducing the likelihood of triggering maternal T cell activation. By limiting classical MHC class I on trophoblasts, the establishes an immune-privileged environment that prevents graft-versus-host-like rejection of fetal tissues. HLA-G exerts immunosuppressive effects by binding to inhibitory receptors such as immunoglobulin-like transcript 2 (ILT2) and ILT4 on immune cells, directly inhibiting the cytotoxic functions of both CTLs and killer () cells at the maternal-fetal interface. Specifically, HLA-G engagement with ILT2 on CD8+ T cells suppresses CTL proliferation and killing activity, while interaction with ILT2 and ILT4 on cells dampens their and release, collectively promoting without compromising broader antiviral defenses. Recent research indicates that uterine killer (uNK) cells are educated by maternal MHC class I molecules, specifically through interaction of the NKG2A receptor with maternal , promoting self-recognition and functional licensing that supports feto-placental development and lowers risk, as evidenced by large-scale genetic studies of over 150,000 pregnancies. Additionally, the soluble isoform of HLA-G (sHLA-G), secreted by trophoblasts, further enhances by inducing the expansion and suppressive activity of regulatory T cells (Tregs), which modulate maternal immune responses to paternal antigens and support sustained immune during . This expression profile represents an evolutionary in placental mammals, where the downregulation of classical MHC class I on reduces alloreactivity and fosters successful by evading maternal adaptive immunity. Dysregulation of this system, such as aberrant expression or altered interactions with maternal receptors, has been linked to increased risk of , a hypertensive disorder characterized by shallow invasion and placental ischemia due to failed . Supporting evidence from mouse models demonstrates that Qa-2, the murine homolog of within the H2 complex, is essential for embryonic implantation and placental development; Qa-2-deficient mice exhibit and higher rates of fetal loss, underscoring its conserved role in maternal-fetal immune accommodation.

PirB-Mediated Neural Plasticity

Major histocompatibility complex (MHC) class I molecules, traditionally known for their role in immune recognition, have been found to play an inhibitory function in neural plasticity through interaction with the paired immunoglobulin-like receptor B (PirB) in the mouse central nervous system. PirB, a receptor expressed on neurons, binds specifically to MHC class I ligands, thereby restricting synaptic remodeling in response to experience-dependent stimuli. This discovery was first reported in a study examining visual cortex development, where MHC class I expression on neurons was shown to signal via PirB to modulate plasticity during critical periods. The mechanism involves MHC class I proteins on neuronal surfaces engaging PirB, which activates downstream signaling pathways that suppress (LTP), a key process in synaptic strengthening. Specifically, PirB engagement recruits phosphatases such as SHP-1 and SHP-2, which dephosphorylate and inhibit MAP kinase pathways essential for , thereby limiting the extent of LTP induction at hippocampal and cortical synapses. In the , this interaction restricts plasticity, the ability of visual cortical neurons to shift preferences based on monocular deprivation during early development. Neurons lacking functional PirB exhibit enhanced LTP and prolonged critical periods for , allowing greater synaptic reorganization even in adulthood. In PirB knockout mice, the absence of this inhibitory signaling leads to structural changes supporting heightened , including increased density and formation in the following sensory perturbations. These findings highlight PirB-MHC class I as a brake on neural adaptability, potentially to stabilize mature circuits after developmental windows close. For human relevance, PirB's ortholog, leukocyte immunoglobulin-like receptor B2 (LILRB2), shares structural and functional similarities, and modulating LILRB2 has been proposed to extend plasticity windows for treating , a disorder of visual development where critical periods are limited. Blocking PirB in adult mice restores plasticity akin to juvenile levels, suggesting therapeutic potential for LILRB2-targeted interventions in humans to enhance recovery without traditional patching methods.

Tissue-Specific Expression

MHC class I molecules, including the classical (HLA) subtypes HLA-A, HLA-B, and , are ubiquitously expressed on the surface of nearly all nucleated cells throughout the body, providing a foundational mechanism for to cytotoxic T cells. This broad distribution ensures continuous immune surveillance against intracellular pathogens and abnormal cells. In contrast, expression is absent on mature erythrocytes due to their anucleate nature, which precludes the synthesis of MHC class I proteins. Similarly, basal MHC class I levels on neurons are characteristically low, reflecting a specialized to minimize immune-mediated interference with neural function while maintaining responsiveness to inflammatory cues. Expression of MHC class I is highly dynamic and inducible, particularly in response to inflammatory signals. Pro-inflammatory cytokines such as interferon-gamma (IFN-γ) and tumor factor-alpha (TNF-α) potently upregulate MHC class I on various cell types, including muscle fibers and endothelial cells, enhancing during infection or tissue damage. This induction occurs through transcriptional activation of MHC class I via cytokine-responsive elements, amplifying immune detection in inflamed tissues. Conversely, many tumors exhibit downregulated MHC class I expression as an immune evasion strategy, often mediated by epigenetic mechanisms involving deacetylases (HDACs) that repress transcription; HDAC inhibitors, such as , can reverse this suppression by promoting and restoring surface expression on tumor cells. Non-classical MHC class I molecules, including , HLA-F, and , display more restricted tissue-specific patterns compared to their classical counterparts. is predominantly expressed on cells at the maternal-fetal interface, contributing to during , with minimal presence in other adult tissues. shows limited distribution, primarily on activated immune cells and , while HLA-F expression is notably enriched in placental tissues, including cytotrophoblasts and syncytiotrophoblasts, where it supports proliferation and . These non-classical molecules often exhibit lower polymorphism and specialized ligand-binding properties tailored to their localized roles.01756-7/fulltext) During development, MHC class I expression undergoes significant changes to balance immune protection and tolerance. In early preimplantation embryos, classical MHC class I levels are low or undetectable, minimizing the risk of maternal immune rejection during the initial stages of . Post-implantation, expression gradually increases in differentiating tissues, particularly in the and fetal organs, coinciding with the establishment of the maternal-fetal interface and the onset of immune competence. This temporal regulation ensures that embryonic cells evade surveillance early on while acquiring the capacity for as development progresses.

Pathogen Interactions

Viral Interference Mechanisms

Viruses have evolved diverse mechanisms to interfere with MHC class I , thereby evading recognition by cytotoxic T lymphocytes (CTLs) and promoting persistent . These strategies target various stages of the MHC class I pathway, including assembly, trafficking, and surface expression, often exploiting host cellular machinery for degradation or retention. Such evasions not only impair adaptive immunity but also alter interactions with innate effectors like natural killer () cells. One prominent approach involves direct inhibition of MHC class I surface expression. The HIV-1 accessory protein Nef downregulates MHC class I molecules by promoting their rapid from the plasma membrane through a - and dynamin-independent pathway involving factor 6 (ARF6), followed by sequestration in the trans-Golgi network or lysosomal degradation. Similarly, human cytomegalovirus (HCMV) encodes US2 and US11 glycoproteins that cause ER retention and dislocation of newly synthesized MHC class I heavy chains to the , where they are targeted for proteasomal degradation via the ER-associated degradation (ERAD) pathway, requiring ubiquitination and interaction with Derlin-1. Another strategy focuses on blocking peptide loading and transport in the ER. Adenovirus type 2 E19 protein binds directly to MHC class I heavy chains in the ER, retaining the complex via a dilysine ER retrieval motif and preventing association with the peptide transporter , thus inhibiting maturation and surface transport. Epstein-Barr virus (EBV) employs the BNLF2a protein, a short hydrophobic , to inhibit function by binding to its nucleotide-binding domain, thereby blocking ATP-dependent peptide translocation into the ER and reducing stable MHC class I- complex formation on the cell surface. Viruses also achieve global suppression of MHC class I expression by disrupting interferon (IFN) signaling pathways that normally upregulate . Measles virus (MV) V protein binds to and STAT2, sequestering them in the and preventing their translocation upon IFN-α/β stimulation, which blocks the induction of IFN-stimulated genes including those enhancing MHC class I transcription and assembly. This suppression not only limits basal MHC class I levels but also prevents IFN-mediated amplification during infection. These interference mechanisms carry significant consequences for host immunity, particularly increasing susceptibility to cell due to diminished inhibitory ligands (e.g., /B/C) on infected cells, activating the "missing self" recognition pathway. However, viruses often counter this by additional evasions, such as HCMV UL18 mimicking MHC class I to engage inhibitory receptors. Over evolutionary timescales, such pressures have driven diversification; for instance, clades exhibit sequence variations in Nef that modulate MHC class I downregulation efficiency, adapting to host HLA polymorphisms and contributing to clade-specific persistence.

Bacterial Modulation Strategies

Bacteria employ diverse strategies to modulate MHC class I , thereby evading cytotoxic T lymphocyte (CTL) recognition and promoting intracellular survival. Intracellular pathogens such as escape from the phagosomal vacuole into the host cytosol using the pore-forming toxin listeriolysin O, allowing replication in a compartment where proteins can be accessed by the proteasomal degradation pathway for MHC class I loading; however, this escape also enables L. monocytogenes to induce type I interferons (IFN-α/β), which sensitize infected macrophages to and suppress IFN-γ-mediated activation, thereby reducing effective CTL responses. Similarly, resides within modified phagosomes that resist fusion with lysosomes and the (), limiting the delivery of phagosomal antigens to the cytosolic proteasomal machinery and ER peptide-loading complex, thereby impairing to + T cells in dendritic cells. Surface-associated modulation occurs in extracellular bacteria like , which adheres to epithelial cells and induces the shedding or of host surface molecules, including and HLA-B, through interactions with CEACAM receptors on antigen-presenting cells; this reduces MHC class I availability for CTL recognition and suppresses activation of antigen-specific + T cells. The host counters these strategies through signaling, particularly IFN-γ, which induces the formation of immunoproteasomes by replacing constitutive subunits with immunosubunits (LMP2, LMP7, and MECL-1), enhancing the generation of peptides suitable for MHC class I binding and improving against intracellular bacteria like L. monocytogenes and M. tuberculosis. This adaptive response restores efficient CTL activation, underscoring the dynamic interplay between bacterial evasion tactics and host immune countermeasures.

Therapeutic Implications for Infections

Therapeutic strategies targeting MHC class I have emerged as promising approaches to bolster + T cell responses against infectious diseases, particularly where pathogens evade immune surveillance by downregulating MHC class I expression. -based vaccines designed to mimic -MHC class I complexes aim to directly stimulate cytotoxic T lymphocytes by delivering pathogen-derived epitopes that bind to patient-specific HLA alleles, thereby enhancing antigen-specific immunity without relying on endogenous processing. For instance, multi-epitope vaccines targeting HIV-1 envelope gp120 have been developed to induce broad neutralizing antibodies and T cell responses, addressing the virus's high variability and immune escape mechanisms. These vaccines, often formulated with adjuvants to promote , have shown potential in preclinical models to restore MHC class I-restricted recognition in chronically infected cells. Similarly, vaccines for (HCV) core and non-structural proteins have demonstrated induction of HLA-A2-restricted + T cells, offering a therapeutic avenue for persistent infections. Immune checkpoint inhibitors, such as anti-PD-1 antibodies, have been investigated to counteract T cell exhaustion in chronic viral infections, where prolonged exposure leads to upregulated PD-1 expression on + T cells, impairing their recognition of MHC class I-presented viral . In (HBV) infection, PD-1 blockade restores the functionality of exhausted HBV-specific + T cells, enhancing viral clearance in preclinical and early clinical trials; for example, nivolumab combined with therapeutic has achieved functional cures in some HBV-positive patients by reinvigorating MHC class I-restricted responses. As of 2025, combination therapies like VTP-300 therapeutic vaccine with nivolumab have shown sustained reductions and functional cures in a subset of chronic HBV patients in phase II trials. This approach leverages the natural role of MHC class I in presenting HBV epitopes to + T cells, countering the virus's partial evasion tactics like altered processing. Clinical trials for chronic HBV and have reported improved T cell proliferation and reduced viral loads with anti-PD-1 therapies, though risks such as HBV reactivation necessitate careful monitoring. Gene therapy strategies focus on restoring MHC class I expression in cells downregulated by viral interference, such as through adenoviral vectors delivering β2-microglobulin (β2M) to reconstitute functional MHC class I complexes on infected cell surfaces. In murine models of viral infection, β2M transfer has successfully upregulated MHC class I, enabling + T cell-mediated of infected targets that would otherwise evade detection. Adaptations of chimeric antigen receptor ( therapy incorporating MHC class I-restricted T cell receptors (TCRs) target intracellular viral antigens presented by MHC class I, with preclinical studies demonstrating efficacy against and by engineering patient-derived T cells to recognize specific pMHC complexes. These TCR-CAR constructs maintain HLA restriction, ensuring precise targeting while overcoming viral downregulation. Recent advances in the 2020s, informed by platforms, have highlighted the potential of mRNA-based therapeutics to enhance MHC class I of viral antigens. s, such as those encoding epitopes optimized for HLA binding, promote efficient cytosolic translation and proteasomal processing in antigen-presenting cells, leading to robust + T cell priming via MHC class I. Insights from these vaccines have spurred designs incorporating MHC class I trafficking signals to boost of exogenous antigens, as seen in experimental mRNA constructs that elicit stronger T cell responses against and other respiratory viruses. This approach not only accelerates development but also translates to therapeutic settings for chronic infections by amplifying MHC class I-dependent immunity.

Genetics and Polymorphism

Human HLA Genes and Loci

The human (MHC) class I genes, known as (HLA) genes, are located within the MHC locus on the short arm of at position 6p21.3. This locus encompasses a densely packed genomic region that includes the classical HLA class I genes HLA-A, HLA-B, and HLA-C, which span approximately 1.5 Mb and play central roles in to cytotoxic T cells. These genes are oriented telomerically, with HLA-A positioned most distally, followed by HLA-H pseudogene, HLA-J pseudogene, HLA-G, HLA-F, HLA-E, and then HLA-C and HLA-B toward the . Each classical HLA class I consists of eight s, encoding a heavy chain that forms a heterodimer with β2-microglobulin. 1 encodes the leader for signal , exons 2 and 3 encode the α1 and α2 extracellular s that form the peptide-binding groove, exon 4 encodes the α3 immunoglobulin-like for interaction, exon 5 encodes the transmembrane region, and exons 6, 7, and 8 encode cytoplasmic domains involved in intracellular signaling and stability. This conserved exon-intron organization across , , and facilitates the structural integrity of the MHC class I molecule, enabling peptide loading and surface expression. Adjacent to the classical genes within the same chromosomal region are three non-classical HLA class I genes: , HLA-F, and , which exhibit limited polymorphism and specialized immunomodulatory functions. primarily presents signal peptides derived from classical MHC class I molecules to inhibit natural killer () cell activity via CD94/NKG2 receptors, contributing to . HLA-F, though less well-characterized, is implicated in interactions with NK cells and T cells during early and viral infections, potentially modulating immune responses through inhibitory signaling. is predominantly expressed at the maternal-fetal interface, where it suppresses T cell and cell to promote allograft tolerance, often via soluble isoforms that bind inhibitory receptors like LILRB1. The immense diversity of HLA class I genes arises from extensive allelic variation, cataloged and standardized by the IPD-IMGT/HLA Database, the official repository for WHO-nominated sequences. As of 2025, this database records over 29,000 alleles for HLA class I genes collectively, with HLA-A encompassing 8,949 alleles, HLA-B 10,680, HLA-C 8,944, HLA-E 385, HLA-F 126, and HLA-G 194; allelic nomenclature follows a systematic format (e.g., HLA-A*02:01:01) denoting gene, allele group, protein, and synonymous variants. This polymorphism, concentrated in exons 2 and 3, underpins population-specific immune repertoires and transplant compatibility.

Non-Human Isotypes and Alleles

In mice, the (MHC) class I region, known as , encodes three classical loci: H2-K, H2-D, and H2-L, which present peptides to + T cells and exhibit haplotype-specific expression, with H2-L absent in certain strains like C57BL/6. Non-classical isotypes in mice include those in the Qa (H2-Q) and Tla (H2-T) regions, comprising over 30 class Ib genes that display tissue-specific expression and limited polymorphism, often involved in specialized immune functions such as cell regulation. These non-classical molecules, like Qa-1, are orthologous to and bind nonamer peptides derived from classical MHC class I signal sequences to modulate immune responses. Among non-human primates, the chimpanzee MHC class I genes are designated Patr-A, Patr-B, and Patr-C, serving as orthologs to human HLA-A, HLA-B, and HLA-C, respectively, with one functional copy per haplotype and an additional Patr-AL lineage in some individuals that resembles HLA-A. In contrast, New World monkeys (Platyrrhini) exhibit expansions of MHC class I lineages, particularly G-like and B-like genes, arising from ancestral duplications that diversified after divergence from Old World monkeys, resulting in multiple paralogs such as Patr-G equivalents in species like the cotton-top tamarin. These expansions contribute to species-specific allelic repertoires adapted to distinct pathogen pressures. Rodent MHC class I systems, exemplified by mice and rats, feature fewer polymorphic alleles at classical loci compared to —typically dozens rather than thousands—but compensate with higher gene copy numbers, including extensive class Ib duplications that enhance functional diversity through multigene families. In birds, classical MHC class I genes are present but often reduced in number and organization compared to mammals; for instance, chickens possess only two dominantly expressed classical loci (BF1 and BF2) within a compact MHC, with passerines showing higher copy numbers and polymorphism in some lineages, though non-classical forms predominate in expression patterns. In pigs, the swine leukocyte antigen (SLA) complex includes three classical MHC class I genes, SLA-1, SLA-2, and SLA-3, which are ubiquitously expressed and polymorphic, playing critical roles in immune recognition. These loci are focal in research, where human antibodies target SLA-1, -2, and -3, prompting strategies like CRISPR-mediated knockouts to generate MHC class I-null pigs that survive without eliciting acute rejection in preclinical models.

Population Diversity and Typing

MHC class I molecules, encoded by (HLA) genes, exhibit extensive polymorphism that varies across human populations, influencing immune responses to pathogens and disease susceptibility. For instance, HLA-B57 alleles, particularly HLA-B57:03, occur at higher frequencies in certain sub-Saharan African populations (approximately 5-10% in groups like South African Blacks) and confer protection against HIV-1 progression by restricting viral replication through cytotoxic T-cell responses. In contrast, *02, the most common HLA-A allele globally, is present at frequencies exceeding 20-50% in diverse populations, including Europeans, Asians, and Africans, contributing to broad presentation capabilities. These allelic distributions are tracked in resources like the Allele Frequency Net Database (AFND), which as of 2025 catalogs over 156,000 HLA allele frequencies from more than 14 million individuals across global populations, enabling comparative analyses of ethnic variations. Such diversity often manifests in haplotypes with strong (LD), where alleles at , -B, and -C loci are inherited together more frequently than expected by chance, as seen in extended MHC blocks spanning hundreds of kilobases that preserve ancestral combinations in human populations. Certain alleles are strongly associated with autoimmune diseases due to their role in . HLA-B*27, for example, is carried by 60-90% of patients with worldwide and increases disease risk up to 100-fold in carriers, likely through aberrant presentation of arthritogenic peptides that trigger inflammation. HLA typing methods have evolved from serological assays, which detect surface antigens using antibodies but are now largely outdated due to limited resolution and inability to distinguish many alleles, to molecular techniques. provides allele-level resolution for exons 2-4 of HLA class I genes but is labor-intensive and costly for large-scale studies. Next-generation sequencing (NGS) has become the gold standard for high-resolution typing, enabling full-length sequencing of HLA loci with accuracy >99% and throughput for population screening, as validated in transplant matching protocols.

Evolutionary Aspects

Gene Duplication and Diversification

The multiplicity of MHC class I genes originated from ancient duplication events in the common ancestor of jawed vertebrates approximately 500 million years ago (Mya), when an ancestral peptide-binding gene underwent tandem duplications to form the proto-MHC cluster. These early duplications established the tandemly arrayed structure of the MHC region, characterized by repeated blocks of class I genes interspersed with non-MHC genes, a pattern conserved across vertebrates from sharks to mammals. This ancestral arrangement provided the genomic foundation for the adaptive immune system's antigen presentation capabilities, with subsequent expansions driven by segmental duplications that increased gene copy number and functional diversity. In humans, the classical MHC class I genes and emerged through segmental duplications of non-classical during early , approximately 40– , while arose later from the HLA-B lineage around 15 . These events involved large-scale genomic rearrangements in the MHC class I region on , where duplicated segments generated paralogous loci that diverged under selective pressures to specialize in presenting diverse peptides to cytotoxic T cells. Non-classical genes like and HLA-F, which arose from earlier duplications, retained more conserved functions, such as roles in cell regulation, whereas emerged more recently; this highlights how duplication facilitated both innovation and specialization within the family. Copy number variation (CNV) further contributes to MHC class I diversification, with some individuals carrying extra copies of pseudogenes such as HLA-H, a non-functional paralog derived from HLA-A. This variability, often resulting from unequal crossing-over in the tandemly repeated MHC region, can expand or contract the total number of class I loci per haplotype, influencing the breadth of the presented peptide repertoire and immune responsiveness. For instance, haplotypes with additional HLA-H copies may indirectly affect adjacent functional genes through linkage, altering antigen presentation diversity without producing viable proteins themselves. Pathogen-driven selection has been a primary force in MHC class I gene diversification, promoting duplications and allelic variation to enhance host survival. arises because individuals with diverse MHC class I alleles can present a wider array of pathogen-derived peptides, evading immune escape by microbes and increasing resistance to . This selective pressure is evident in higher MHC diversity in pathogen-rich environments, where duplication events amplify the genomic substrate for polymorphism, ensuring broader immune coverage across populations.

Birth-and-Death Evolution Model

The birth-and-death evolution model posits that multigene families such as MHC class I evolve through a dynamic process of , which generates new genes ("birth"), followed by and potential loss via disabling leading to pseudogenization or deletion ("death"). Proposed by Nei and Hughes in , this model accounts for the observed polymorphism and turnover in MHC loci, where duplicate genes may either become fixed through positive selection or be eliminated if they confer no advantage or are deleterious. Under this framework, the functional repertoire of MHC class I molecules is maintained despite high rates of gene gain and loss, allowing adaptation to diverse pathogens without concerted homogenization across family members. Evidence supporting the model in MHC class I comes from phylogenetic analyses of lineages, where a substantial proportion of sequences represent pseudogenes, reflecting frequent death. For instance, in primates like the owl monkey, numerous MHC class I pseudogenes cluster with functional loci, indicating recent turnover events that align with the birth-and-death process. Additionally, positive Darwinian selection drives rapid evolution at peptide-contact residues in the antigen-binding groove, as evidenced by dN/dS ratios exceeding 1 in these sites across HLA class I loci, promoting diversification of presentation capabilities. Balancing selection plays a key role in preserving polymorphisms under this model, with heterozygote advantage enabling individuals to recognize and respond to a wider array of -derived peptides compared to homozygotes. Pathogen-driven pressures further favor rare alleles through negative , where less common variants evade prevalent pathogen evasion strategies, thereby sustaining allelic over time. In humans, the substitution rate at MHC class I loci approximates 10^{-9} per site per year, underscoring the relatively rapid allelic turnover consistent with birth-and-death dynamics. This rate, combined with duplication events, facilitates ongoing of the MHC class I family to counter shifting selective pressures from pathogens.

Comparative Phylogeny Across Species

MHC class I molecules are conserved across all vertebrates (gnathostomes), where they play a central role in to cytotoxic T cells as part of the . This conservation extends from cartilaginous like , which possess tightly linked class I genes with antigen-processing components such as and LMP, to higher vertebrates. Recent studies have identified a primitive W-category of MHC molecules in cartilaginous , exhibiting features of both class I and II, suggesting an ancestral form from which modern class I diverged. In contrast, jawless vertebrates (agnathans), including lampreys and , lack MHC class I and II genes entirely, relying instead on alternative adaptive immunity mechanisms like variable receptors (VLRs) for . In mammals, MHC class I gene repertoires exhibit significant expansions in certain lineages, particularly among . For instance, (Bos taurus) harbor at least six classical class I loci (BoLA-1 through BoLA-6) alongside ten non-classical loci, totaling over ten class I genes per , which supports diverse presentation tailored to pressures in ruminants. Conversely, monotremes such as the (Ornithorhynchus anatinus) show contractions in their MHC class I repertoire, with classical class Ia genes colocalizing with class II and antigen-processing genes in a more compact arrangement reminiscent of non-mammalian vertebrates, reflecting an ancestral configuration with fewer duplicated loci. Non-mammalian vertebrates display varied MHC class I architectures shaped by lineage-specific duplications. In amphibians like (African clawed frog), the class I region is minimal, featuring a single classical class Ia locus closely linked to and LMP genes, with additional non-classical class Ib genes located more distantly. Teleost fish, following their ancient whole-genome duplication approximately 350 million years ago, exhibit duplicated MHC class I genes, with classical loci often linked to antigen-processing machinery but unlinked from class II, enabling independent evolution and enhanced diversity in response to aquatic pathogens. The ancient origins of MHC class I trace back to a duplication event from an ancestral class II-like gene around 450 million years ago, coinciding with the emergence of adaptive immunity in vertebrates and subsequent of immune gene families. This duplication established the foundational class I/II linkage observed in basal gnathostomes, with subsequent birth-and-death evolutionary dynamics driving species-specific diversification.