Fact-checked by Grok 2 weeks ago

Integrin

Integrins are a large family of heterodimeric transmembrane receptors that mediate to the () and to other cells, facilitating essential interactions in multicellular organisms. These receptors consist of non-covalently associated α and β subunits, each featuring an extracellular domain for binding, a single transmembrane , and a short cytoplasmic tail that connects to the . In humans, 18 α subunits and 8 β subunits combine to form at least 24 distinct integrin heterodimers, each with specific preferences and tissue distributions. Integrins were first identified in the 1970s as receptors mediating platelet adhesion, with the broader family characterized in the 1980s through work by researchers including Richard Hynes, Erkki Ruoslahti, and Timothy A. Springer, who were awarded the Lasker~DeBakey Clinical Medical Research Award in 2022 for their discoveries. The extracellular portion of integrins adopts a head-to-tail arrangement, with the α subunit often containing a seven-bladed β-propeller domain and, in some cases, an inserted I-domain for ligand recognition, while the β subunit includes an I-like domain critical for activation. Activation of integrins involves conformational changes from a low-affinity bent state to a high-affinity extended state, regulated by intracellular signals (inside-out signaling) or extracellular ligands (outside-in signaling), which enable divalent cation-dependent binding to ECM components like fibronectin, collagen, and laminin, or to counter-receptors on adjacent cells. This bidirectional signaling integrates mechanical forces and biochemical cues, linking the ECM to intracellular pathways that control cytoskeletal dynamics. Integrins play pivotal roles in fundamental biological processes, including embryonic development, , immune responses, and tissue homeostasis, by promoting , , survival, and . Dysregulation of integrin is implicated in various pathologies, such as cancer metastasis, , and autoimmune diseases, underscoring their therapeutic potential. Evolutionarily conserved across metazoans for over 600 million years, integrins represent a of cellular and intercellular communication.

Introduction

Definition and General Role

Integrins are heterodimeric transmembrane receptors composed of non-covalently associated α and β subunits that mediate to the () as well as cell-cell interactions. These receptors function as bidirectional signaling molecules, enabling inside-out signaling where intracellular cues regulate binding affinity and outside-in signaling where extracellular engagement triggers intracellular pathways. Integrins play essential roles in fundamental cellular processes, including , , , and , by linking the or adjacent cells to the actin cytoskeleton and modulating signaling cascades such as those involving focal adhesion kinase and Rho GTPases. For instance, they facilitate cell spreading on ECM components like through heterodimer formation on the cell surface. The integrin family is evolutionarily conserved across multicellular animals, from like sea urchins to mammals, underscoring their ancient origin in metazoan adhesion mechanisms. In humans, 18 α subunits and 8 β subunits combine to form 24 distinct heterodimers, each with specific specificities and distributions. Due to their central involvement in inflammatory and immune responses, integrins represent key therapeutic targets; for example, the natalizumab inhibits α4 integrins to block leukocyte migration into the , providing efficacy in treating .

Discovery and Historical Context

The discovery of integrins began in the mid-1970s amid investigations into and the , particularly the role of in attachment. In 1976, Richard O. Hynes identified fibronectin receptors on the surface of transformed , linking the protein's distribution to intracellular filaments through studies, which suggested a transmembrane connection mediating cell-matrix interactions. This work built on earlier observations of (then called LETS protein) loss in malignant cells and laid the groundwork for recognizing receptors. Key milestones in the 1980s advanced the molecular characterization of these receptors. In 1984, Erkki Ruoslahti and Michael D. Pierschbacher identified the Arg-Gly-Asp (RGD) tripeptide sequence within as the critical motif for cell attachment, demonstrating that synthetic peptides containing RGD could mimic fibronectin's adhesive function. This was followed in by the purification of the α5β1 integrin, the primary receptor, from human osteosarcoma cells using on a column coupled to the cell-binding fragment of , confirming its ligand-binding properties. The heterodimeric structure was further characterized in subsequent studies. Early functional studies in the same decade employed monoclonal antibodies to block cell attachment; for instance, antibody (later termed J1H) against the β1 subunit inhibited spreading on , establishing integrins' direct role in . The late 1980s saw rapid progress in genetic characterization, with cDNA cloning of integrin subunits revealing their conserved structure and diversity. The β1 subunit was cloned in 1986 from chicken embryo fibroblasts, showing to other molecules, while human α5 and other α subunits followed in 1987, enabling and confirmation of the integrin family. In 1987, Hynes coined the term "integrin" to describe this superfamily of transmembrane receptors that integrate the with extracellular ligands. Nomenclature evolved alongside these discoveries, transitioning from ad hoc designations to a systematic framework. Integrins were initially termed "very late antigens" (VLA-1 through VLA-6) in based on their delayed expression on activated T lymphocytes, referring primarily to β1-containing heterodimers. By the early , with accumulating structural data, the community adopted the standardized αβ notation (e.g., α5β1 for the receptor), as proposed in comprehensive reviews, facilitating classification of the growing family.

Molecular Structure

Subunit Composition and Assembly

Integrins are transmembrane heterodimeric receptors composed of non-covalently associated α and β subunits, forming a 1:1 essential for their function. In humans, there are 18 distinct α subunits and 8 β subunits, which can pair to generate at least different integrin heterodimers, with each α subunit typically determining the ligand-binding specificity while β subunits are shared among multiple α partners. The extracellular domains of the α and β subunits dimerize in a head-to-head , creating a ligand-binding head region, while their transmembrane and cytoplasmic domains extend like legs to connect the to the . The assembly of integrin heterodimers occurs co-translationally in the (), where α and β subunits must pair correctly for proper folding, , and trafficking to the plasma membrane. Unpaired α or β subunits are retained in the and targeted for via the ER-associated (ERAD) pathway, ensuring that only functional heterodimers reach the surface. This quality control mechanism prevents the accumulation of misfolded or incomplete integrins, maintaining cellular . For instance, excess β4 subunits, if not paired with α6, are rapidly degraded in the . Certain β subunits exhibit variations in structure that influence their assembly and downstream functions. Notably, the β8 subunit lacks a typical cytoplasmic tail, including motifs like NPXY that are present in other β subunits and crucial for intracellular signaling; this feature results in αvβ8 integrins that primarily mediate presentation rather than direct . Such structural differences highlight the diversity in subunit composition that allows integrins to adapt to specific cellular contexts.

Domain Organization

Integrins are heterodimeric transmembrane receptors composed of α and β subunits, each featuring a modular architecture that spans the , , and intracellular milieu. The extracellular region of the α subunit consists of a seven-bladed β-propeller at the , which serves as the primary for interaction through its upper face, followed by a (an immunoglobulin-like β-sandwich of 140–170 residues), and two calf domains (calf-1 and calf-2, also β-sandwich folds) that form the lower leg of the subunit. Approximately half of the α subunits additionally contain an inserted I- (~200 residues) between blades 2 and 3 of the β-propeller, which harbors a metal ion-dependent (MIDAS) for binding. In contrast, the β subunit's extracellular includes an () with an α/β fold, an inserted (β-sandwich), a βI (also known as the I-like ) containing a MIDAS motif for metal ion coordination, four cysteine-rich EGF-like repeats, and a flexible β-tail . The transmembrane domains of both α and β subunits are single-span α-helices, each comprising approximately 20–25 residues, which facilitate non-covalent dimerization and the transmission of mechanical forces across the . These helices exhibit specific orientations, with the α helix often perpendicular to the membrane and the β helix tilted, contributing to the overall stability of the heterodimer. Cytoplasmic domains are short, unstructured tails of 20–50 residues that extend into the , enabling interactions with the and intracellular signaling proteins; the β subunit tail notably features conserved NPxY motifs that serve as binding sites for adaptor proteins. Recent advances in cryo-electron microscopy (cryo-EM) have provided high-resolution models of full-length integrins, such as αIIbβ3, at resolutions below 4 (e.g., 3.1–3.4 for apo and ligand-bound states in native as of 2023), revealing the detailed arrangement of all 12 extracellular subdomains in bent conformations where the head (β-propeller and βI domains) is angled relative to the legs (, , and EGF domains), with separated transmembrane helices and no observable cytoplasmic density due to flexibility. More recent studies as of 2025 have achieved even higher resolutions, such as 2.67–2.85 for αIIbβ3 in multiple conformations from native platelet membranes, and structures of other integrins like αEβ7 in apo and ligand-bound states, further elucidating conformational dynamics and interactions without altering the core domain folds. These structures confirm the modular organization and highlight conformational flexibility between bent and extended states across inactive and active forms.

Activation and Regulation

Conformational Changes

Integrins exist predominantly in a bent, low-affinity conformation in their inactive state, characterized by a compact where the headpiece (comprising the β-propeller and β-I-like domains) folds back toward the membrane-proximal legs (thigh, calf-1, and calf-2 domains in the α-subunit, and thigh and calf-1 in the β-subunit). This bent form, observed in physiological conditions with Ca²⁺/Mg²⁺ ions, exhibits minimal binding, as the headpiece is positioned too close to the membrane for effective interaction. Upon , integrins transition to an extended, high-affinity conformation through a switchblade-like mechanism, involving separation of the headpiece from the legs and splaying of the leg domains away from each other. In this extended state, induced by Mn²⁺ or high-affinity ligands like cyclo-RGDfV, the integrin achieves greater than 98% extension, enabling robust adhesion to ligands such as fibrinogen or . The pivotal intracellular trigger for this transition is the separation of the α- and β-subunit transmembrane helices, which disrupts their inhibitory association and propagates conformational changes extracellularly. This separation is driven by binding of the talin FERM domain (specifically the F3 subdomain) to the β-integrin cytoplasmic tail, which forms a salt bridge with a conserved aspartate residue (e.g., β3 D723), destabilizing the α-β salt bridge (e.g., αIIb R995-β3 D723) that maintains the inactive state. Concurrently, the talin F2 subdomain's positively charged patch interacts with the membrane, reorienting the β-transmembrane helix by approximately 20°, facilitating helix separation and initiating inside-out signaling. A critical extracellular event in is the outward swing of the hybrid relative to the β-I-like , which unlocks the -binding site. In the bent conformation, the hybrid is clasped against the β-I-like , restraining the α7 and maintaining a low-affinity pose; induces a ~60° swing, pulling downward on the α7 and allowing upward movement of the α1 in the β-I . This motion, linked to the extended leg conformation, enables the I- (in α-subunits) or β-I to adopt an open state for engagement. These structural shifts are allosterically regulated through changes in cation coordination at the metal ion-dependent adhesion site () within the β-I or α-I domain. In the low-affinity , the MIDAS-bound Mg²⁺ or Ca²⁺ ion is coordinated in a manner that limits interaction with aspartate (e.g., in RGD motifs); repositions coordinating residues, enhancing Mg²⁺ for the ligand's carboxylate group and forming a direct (e.g., 3.4 Å to the β1-α1 ). This reconfiguration, propagated from transmembrane separation, increases ligand-binding by orders of magnitude, stabilizing the high-affinity extended form.

Regulatory Mechanisms

Integrin is primarily regulated through inside-out signaling, where intracellular cues trigger conformational changes that increase . In this , talin binds to the β-subunit cytoplasmic tails, recruiting integrins to specific plasma membrane sites and inducing their high- state for ligands. Kindlin cooperates with talin by binding to the same β-tails, enhancing talin and stabilizing the activated conformation to promote potent integrin and cell adhesion.30202-2) The scaffold protein RIAM facilitates this by interacting with activated Rap1 , which recruits talin to the membrane and unfolds its autoinhibitory structure to enable integrin engagement. Negative regulation maintains integrins in a low-affinity state to prevent aberrant . Proteins such as SHARPIN and ICAP-1 inhibit talin to β-tails, thereby suppressing and promoting integrin or . Filamin competes directly with talin for to the β-cytoplasmic tails, stabilizing the inactive integrin conformation and linking it to the in a manner that dampens signals.00678-2) Outside-in feedback loops reinforce integrin following initial engagement. to the extracellular domain transmits signals intracellularly, activating kinases and focal kinase (FAK), which in turn phosphorylate components that sustain the high-affinity state and promote further talin and kindlin recruitment.00242-5) Post-translational modifications fine-tune integrin affinity and localization. Phosphorylation of β-tails, such as by (PKC), modulates binding sites for regulatory proteins, thereby altering affinity and influencing activation dynamics.00238-4) Glycosylation, particularly N-linked modifications on integrin subunits, regulates trafficking from the to the plasma membrane, ensuring proper surface expression and functional maturation.

Core Functions

Cell Adhesion

Integrins serve as primary mediators of , enabling physical connections between cells and the (ECM) or adjacent cells through specific -binding interactions. These transmembrane receptors, composed of α and β subunits, recognize distinct motifs on ECM proteins and counter-receptors, facilitating stable attachments essential for tissue integrity and cellular organization. Upon ligand engagement, integrins cluster to form adhesion structures that link the ECM to the intracellular , thereby transmitting mechanical forces and supporting cellular processes. In adhesion to the ECM, integrins bind to key structural components such as , , , and . The α5β1 integrin specifically interacts with via the arginine-glycine-aspartic acid (RGD) motif, promoting and endothelial cell attachment. Similarly, α1β1 and α2β1 integrins recognize types I and IV, enabling adhesion in connective tissues and basement membranes. Laminin binding is mediated by α3β1 and α6β1 integrins, which are crucial for epithelial cell anchorage, while αvβ3 integrin engages through RGD-dependent interactions, supporting endothelial and cell adhesion in vascular contexts. Cell-cell involving integrins occurs prominently in immune responses, where the leukocyte-specific αLβ2 (LFA-1) integrin binds to intercellular molecule-1 () on endothelial cells, facilitating and firm arrest under shear flow. This interaction is vital for recruiting immune cells to sites. Upon engagement, integrins cluster into , dynamic multiprotein complexes that anchor cells to the substrate. These structures incorporate intracellular proteins such as and paxillin, which connect integrin tails to the , stabilizing adhesions and distributing mechanical loads. formation begins with integrin diffusion and clustering into puncta, progressing to mature plaques that reinforce attachment. A key feature of integrin-mediated is force-dependent reinforcement through catch bonds, where applied tensile force prolongs the bond lifetime rather than dissociating it. For instance, the α5β1-fibronectin interaction exhibits catch bond behavior, allowing adhesions to strengthen under physiological shear or traction forces, thus enhancing cellular stability. These adhesions can also initiate signaling cascades, though the biochemical outputs are detailed elsewhere.

Upon ligand binding to the extracellular domain of integrins, outside-in signaling is initiated, leading to the activation of intracellular kinases such as focal adhesion kinase (FAK) and family kinases. This activation occurs through autophosphorylation of FAK at tyrosine 397, which serves as a docking site for , forming a FAK- complex that propagates downstream signals. These events trigger multiple pathways, including the /extracellular signal-regulated kinase (MAPK/ERK) cascade, which promotes cell proliferation, and the / (PI3K/Akt) pathway, which enhances cell survival and inhibits . A key mediator in this signaling is integrin-linked kinase (ILK), which assembles into a heterotrimeric complex with particularly interesting new Cys-His protein (PINCH) and parvin to bridge integrins with the . This ILK-PINCH-parvin () complex facilitates by regulating Rho family , such as RhoA, Rac1, and Cdc42, which control cytoskeletal dynamics and cell motility. Although ILK possesses pseudokinase activity, its primary role in the IPP complex is structural, enabling the recruitment of effectors that modulate activity and sustain signaling from focal adhesions. Integrins also engage in crosstalk with growth factor receptors, exemplified by (), where ligand-induced integrin clustering enhances EGFR and amplifies downstream signaling for cooperative regulation of and invasion. This bidirectional interaction allows integrins to modulate EGFR signaling intensity, often through shared activation of FAK and , leading to enhanced MAPK and PI3K pathways in response to extracellular cues. The strength of integrin-mediated signaling is quantitatively influenced by avidity modulation, where receptor clustering increases the effective to in a dose-dependent manner, thereby amplifying downstream activation and pathway outputs. For instance, higher through clustering can shift the dose-response curve for FAK , enabling graded signaling responses proportional to ligand density. This mechanism ensures that signaling scales with strength without altering individual integrin states.

Physiological and Pathological Roles

Development and Tissue Repair

Integrins play crucial roles in embryonic development, particularly during embryogenesis where they mediate cell-ECM interactions essential for morphogenetic processes. The α5β1 integrin is vital for and somitogenesis, facilitating adhesion to that drives axis elongation and segmentation in the developing . Specifically, in models, disruption of α5β1-mediated fibronectin binding leads to arrested axis elongation around embryonic day 9.0, halting somitogenesis due to impaired convergent extension movements. Furthermore, global of the β1 integrin subunit in mice results in embryonic lethality during early postimplantation stages, primarily due to failure of the to develop properly and implantation defects, underscoring β1's indispensable role in early embryonic adhesion and survival. In angiogenesis, integrins support the formation of new blood vessels during development and physiological remodeling. The αvβ3 integrin is highly expressed on endothelial cells and is required for (VEGF)-induced , where it promotes endothelial cell migration, , and tube formation by integrating signals from the . Antagonists targeting αvβ3 have been developed as anti-angiogenic therapies, demonstrating inhibition of endothelial and vessel maturation in preclinical models, highlighting its therapeutic potential in modulating pathological while preserving developmental processes. During tissue repair, integrins orchestrate cellular responses to injury, including remodeling and . In wound healing, the α2β1 integrin on fibroblasts binds fibrils, enabling their contraction and reorganization to form and restore tissue integrity; mice lacking α2β1 exhibit reduced formation and diminished wound strength, indicating its critical function in remodeling post-injury. Similarly, in peripheral nerve regeneration, β1 integrins, including α1β1, facilitate migration along -rich scaffolds, supporting axonal regrowth and remyelination after nerve damage. Integrins also contribute to maintaining stem cell niches, ensuring self-renewal and differentiation potential. In the hematopoietic stem cell (HSC) niche within the , β1 integrins mediate adhesion to components like and , which is essential for HSC retention, quiescence, and long-term repopulation capacity; disruption of β1 function impairs HSC homing and niche occupancy, leading to defective hematopoiesis. This interaction supports the maintenance of HSC stemness, analogous to pluripotency regulation in other contexts, through signaling pathways that preserve undifferentiated states.

Disease Associations

Integrins play a central role in numerous pathological processes, particularly through their dysregulation in , where specific subtypes facilitate tumor progression. The αvβ3 and αvβ5 integrins are highly expressed in tumor vasculature and promote by mediating endothelial to the and supporting signaling. In , the α6β4 integrin enhances and by linking to the and activating pro-migratory pathways such as PI3K/Akt. Therapeutic targeting of these integrins, such as with the RGD-mimetic cilengitide, which inhibits αvβ3 and αvβ5, showed promise in preclinical models but failed in Phase III trials for due to lack of overall survival benefit, highlighting challenges in integrin antagonism for . In autoimmune and inflammatory diseases, integrins mediate leukocyte trafficking and contribute to chronic inflammation. The α4β7 integrin facilitates gut-homing of T cells in (IBD), and its blockade by the , approved in 2014, induces clinical remission in and by preventing α4β7 binding to MAdCAM-1 on endothelial cells. Similarly, LFA-1 (αLβ2 integrin) drives T-cell in , but the anti-LFA-1 efalizumab, initially approved for moderate-to-severe , was withdrawn in 2009 due to risks of . Fibrosis involves excessive extracellular matrix deposition, where integrin overexpression sustains myofibroblast activation. β1 integrin is overexpressed in lung and kidney fibrosis, promoting epithelial-to-mesenchymal transition and fibroblast proliferation via TGF-β signaling; conditional genetic knockout of β1 in mouse models reduces renal cystogenesis and interstitial fibrosis in polycystic kidney disease. Recent studies (2020s) have focused on αvβ6 integrin in idiopathic pulmonary fibrosis (IPF), where it activates latent TGF-β1 to drive fibrotic remodeling; inhibitors like bexotegrast, a dual αvβ6/αvβ1 antagonist, showed dose-dependent reduction in lung fibrosis markers in early Phase II trials, but its Phase 2b/3 trial was discontinued in 2025 due to safety concerns including increased IPF-related adverse events. Other diseases linked to integrin defects include , a rare inherited bleeding disorder caused by mutations in αIIbβ3 (ITGA2B/ITGB3), leading to impaired platelet aggregation and fibrinogen binding. Emerging research post-2020 implicates αvβ3 in COVID-19-associated vascular complications, where exploits this integrin on endothelial cells to induce dysregulation and . Therapeutic strategies targeting integrins encompass monoclonal antibodies, small-molecule RGD mimetics, and gene-editing approaches. Beyond , other mAbs like (anti-α4 integrin) are used in , though with boxed warnings for PML risk. CRISPR studies reveal knockout phenotypes underscoring therapeutic potential; for instance, α9 integrin depletion in models suppresses by promoting β-catenin degradation. These advances inform ongoing trials, emphasizing combination therapies to overcome resistance observed in early integrin inhibitors, with recent focus (as of 2025) on next-generation αv inhibitors like BG00011 for IPF following discontinuations such as bexotegrast.

Diversity Across Organisms

Vertebrate Integrins

In vertebrates, integrins display a high degree of diversity, primarily through the combinatorial assembly of 18 distinct α subunits and 8 β subunits, which generate 24 unique αβ heterodimers in humans and other mammals. This repertoire enables specialized functions tailored to tissue-specific needs, with expression patterns varying across cell types such as epithelial cells, fibroblasts, and hematopoietic cells. The α subunits are categorized into subfamilies based on their ligand-binding preferences; for instance, the collagen-binding group includes α1, α2, α10, and α11, which primarily pair with β1 to interact with components like collagens. Other prominent α subunits encompass αV, which binds RGD motifs in ligands such as and , often forming complexes like αVβ3 or αVβ5, and αIIb, which is essential for platelet aggregation as part of the αIIbβ3 heterodimer that recognizes fibrinogen. The β subunits further contribute to this specificity, with β1 being the most ubiquitous and versatile, associating with at least 12 different α subunits and expressed on nearly all vertebrate cell types to support broad adhesion roles. In contrast, β2 is leukocyte-specific, forming integrins like αLβ2 (LFA-1) and αMβ2 (Mac-1) that mediate immune cell recruitment and interactions with endothelial cells. β3 is enriched in endothelial cells and platelets, where it participates in and through pairings such as αVβ3 and αIIbβ3, while β8 is notable for its role in activating latent transforming growth factor-β (TGF-β), particularly in epithelial and neuronal contexts via αVβ8. The remaining β subunits (β4, β5, β6, and β7) exhibit more restricted distributions, such as β4 in epithelial hemidesmosomes and β7 in mucosal lymphocytes, enhancing localized functions. This combinatorial diversity results in 24 functional heterodimers, each with distinct tissue expression; for example, β2-containing integrins are predominantly restricted to leukocytes, underscoring their role in immunity, whereas β1 integrins predominate in mesenchymal tissues. Evolutionarily, integrins are highly conserved, with two major α subunit families tracing back to before the deuterostome-protostome split, ensuring core mechanisms across . Subunit expansions, particularly in α and β lineages, occurred in mammals, increasing heterodimer variety compared to earlier vertebrates. In , orthologous integrins like αVβ3 support immune responses, as demonstrated in teleosts such as , where they facilitate leukocyte function and .

Non-Vertebrate Integrins

Integrins in non-vertebrate organisms exhibit structural and functional conservation with their counterparts while displaying reduced diversity in subunit composition, reflecting evolutionary adaptations to simpler multicellular architectures. In model such as the Drosophila melanogaster and the Caenorhabditis elegans, integrins mediate essential processes like muscle attachment and tissue morphogenesis, often with fewer heterodimeric combinations than the 24 found in mammals. In , the integrin repertoire includes five α subunits (αPS1 through αPS5) and two β subunits (βPS, encoded by the myospheroid gene, and βν, encoded by inflated), forming key heterodimers such as PS1 (αPS1/βν) and PS2 (αPS2/βPS). These integrins are critical for embryonic , particularly in establishing myotendinous junctions where the αPS2/βPS complex anchors muscle fibers to the (), analogous to vertebrate focal adhesions and enabling force transmission during contraction. In C. elegans, integrins are even more streamlined, with two α subunits (PAT-2 and INA-1) pairing exclusively with a single β subunit (PAT-3) to form αPAT-2/βPAT-3 and αINA-1/βPAT-3 heterodimers. The αPAT-2/βPAT-3 integrin supports body wall muscle adhesion to the , while αINA-1/βPAT-3 contributes to by facilitating epithelial cell- interactions during organ formation. Structurally, non-vertebrate integrins maintain the canonical αβ heterodimeric architecture with extracellular ligand-binding domains, transmembrane regions, and cytoplasmic tails that link to the actin cytoskeleton via proteins like talin, but they feature notable simplifications compared to s. α subunits, such as those in and C. elegans, lack the inserted I-domain present in many α subunits. Ligand recognition occurs primarily through the β-propeller domain of the α subunit and the I-like domain of the β subunit, which contributes to their specificity for components like and RGD motifs. β subunits in these organisms exhibit shorter cytoplasmic tails compared to many β subunits, with conserved motifs for cytoskeletal linkage via proteins like talin but fewer regulatory sites, potentially limiting bidirectional signaling complexity. These features underscore a more basal role in adhesion rather than the multifaceted signaling seen in s. Evolutionary analyses reveal that integrin-like adhesion systems predate the bilaterian radiation, with proto-integrin components emerging in the last common ancestor of , as evidenced by homologs of integrin-associated proteins (e.g., talin and kindlin) in choanoflagellates like Monosiga brevicollis, though full heterodimeric integrins are a metazoan . In basal metazoans such as cnidarians and platyhelminths, integrins support fundamental adhesion for tissue integrity, diverging into the specialized forms observed in protostomes like arthropods and nematodes well before vertebrate evolution. This conservation of core adhesive functions across non-vertebrates highlights integrins' ancient role in enabling multicellularity through cell-matrix interactions.

References

  1. [1]
    Integrins: An Overview of Structural and Functional Aspects - NCBI
    Integrins are heterodimeric transmembrane receptors that mediate cell-adhesion. With their extracellular head region, most integrins bind extracellular matrix ( ...Introduction · Structural Aspects of Integrins · Integrin-Containing Adhesions
  2. [2]
    Integrin Structure, Activation, and Interactions - PMC - PubMed Central
    Integrins are large, membrane-spanning, heterodimeric proteins that are essential for a metazoan existence. All members of the integrin family adopt a shape ...
  3. [3]
    Targeting integrin pathways: mechanisms and advances in therapy
    Jan 2, 2023 · Integrins are considered the main cell-adhesion transmembrane receptors that play multifaceted roles as extracellular matrix ...
  4. [4]
    Integrins: Bidirectional, Allosteric Signaling Machines - ScienceDirect
    In addition to mediating cell adhesion, integrins make transmembrane connections to the cytoskeleton and activate many intracellular signaling pathways. Since ...
  5. [5]
    Integrins - Molecular Biology of the Cell - NCBI Bookshelf - NIH
    Integrins are the principal receptors used by animal cells to bind to the extracellular matrix. They are heterodimers and function as transmembrane linkers.
  6. [6]
    Integrins during evolution: Evolutionary trees and model organisms
    The integrins form a large family of cell adhesion receptors. All multicellular animals express integrins, indicating that the family evolved relatively ...
  7. [7]
    The integrins | Genome Biology | Full Text - BioMed Central
    Jun 1, 2007 · They are transmembrane αβ heterodimers and at least 18 α and eight β subunits are known in humans, generating 24 heterodimers.
  8. [8]
    Natalizumab: targeting alpha4-integrins in multiple sclerosis - PubMed
    The humanized anti-alpha(4)-integrin antibody natalizumab demonstrated an unequivocal therapeutic effect in preventing relapses and slowing down the pace of ...
  9. [9]
    The emergence of integrins: a personal and historical perspective
    By 1976 significant circumstantial evidence had accumulated that suggested some relationship between extracellular fibronectin-containing fibrils and ...
  10. [10]
    Cell attachment activity of fibronectin can be duplicated by ... - PubMed
    Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature. 1984 May;309(5963):30-3. doi: 10.1038/309030a0.Missing: RGD | Show results with:RGD
  11. [11]
    Integrins: a family of cell surface receptors - PubMed
    1987 Feb 27;48(4):549-54. doi: 10.1016/0092-8674(87)90233-9. Author. R O Hynes. PMID: 3028640; DOI: 10.1016/0092-8674(87)90233-9. No abstract available ...
  12. [12]
    Overview-studying integrins in vivo - PMC - PubMed Central - NIH
    ... unpaired α or β chains are retained in the endoplasmic reticulum and degraded. ... Interestingly, deletion of the α2β1 integrin or the α3β1 integrin alone ...
  13. [13]
    Cell cycle and adhesion defects in mice carrying a targeted deletion ...
    If the biosynthesis of β4 exceeds that of α6, the fraction of unpaired β4 is degraded rapidly in the endoplasmic reticulum (ER) (Giancotti et al., 1992). In ...
  14. [14]
    αvβ8 integrin adhesion and signaling pathways in development ...
    As noted above, the amino acid sequence of the β8 integrin cytoplasmic tail is not homologous to that of other β subunits. Most integrin β subunits ...
  15. [15]
    Cryo-EM structures of full-length integrin αIIbβ3 in native lipids - PMC
    Jul 13, 2023 · We now report the cryo-EM structures of full-length αIIbβ3 in its apo and eptifibatide-bound states in native cell-membrane nanoparticles at near-atomic ...
  16. [16]
    Full-length αIIbβ3 CryoEM structure reveals intact integrin initiate ...
    Feb 24, 2023 · Based on the domain organization, the whole integrin structure could also be split into three regions: Head, Leg, and TM. The integrin ...
  17. [17]
  18. [18]
  19. [19]
    Mechanism of integrin activation by talin and its cooperation with ...
    Apr 29, 2022 · Talin activation allows talin-H binding to the integrin-β CT15,16,17,18, which in turn disrupts the integrin-α/β CT association to trigger the ...<|control11|><|separator|>
  20. [20]
    Conformational activation of talin by RIAM triggers integrin-mediated ...
    Dec 18, 2014 · The structure of an integrin/talin complex reveals the basis of inside-out signal transduction. ... recruitment of talin by RIAM in integrin ...
  21. [21]
    The importance of N-glycosylation on β 3 integrin ligand binding and ...
    Jul 5, 2017 · N-glycosylations can regulate the adhesive function of integrins. Great variations in both the number and distribution of N-glycosylation ...
  22. [22]
    Genetic and cell biological analysis of integrin outside-in signaling
    This review discusses recent work that has tremendously broadened our understanding of the complexity of integrin-mediated signaling.Missing: paper | Show results with:paper
  23. [23]
    Integrin signaling in cancer: bidirectional mechanisms and ...
    Sep 28, 2023 · This review provides an overview of integrin-dependent bidirectional signaling mechanisms in cancer that can either support or oppose tumorigenesis.Missing: paper | Show results with:paper
  24. [24]
    ILK, PINCH and parvin: the tIPP of integrin signalling - Nature
    Dec 21, 2005 · The ternary complex of integrin-linked kinase (ILK), PINCH and parvin functions as a signalling platform for integrins by interfacing with the actin ...Missing: linked kinase
  25. [25]
    The ILK/PINCH/parvin complex: the kinase is dead, long live the ...
    Dec 24, 2009 · The integrin-linked kinase (ILK)/PINCH/parvin (IPP) complex is a central constituent of at least β1 and β3 integrin containing adhesion sites, ...
  26. [26]
    Integrin-linked kinase (ILK) and its interactors | Journal of Cell Biology
    In this review, we discuss the molecular activities and cellular functions of ILK, a protein that is emerging as a key component of the cell–ECM adhesion ...
  27. [27]
    Integrin and Growth Factor Receptor Crosstalk | Circulation Research
    This review will focus on the biological importance of integrin and growth factor receptor crosstalk that occurs during cell growth, migration, and invasion ...
  28. [28]
    The integrin—growth factor receptor duet - Wiley Online Library
    Sep 20, 2007 · Our review describes emerging concepts regarding the cross-talk between integrins and GFRs and highlights the role of integrins in contributing ...Abstract · Mechanisms Of Integrin--Gfr... · Are Integrins Upstream...<|control11|><|separator|>
  29. [29]
    Complementary Roles for Receptor Clustering and Conformational ...
    The effects of integrin clustering and affinity modulation were additive, and clustering promoted irreversible ligand binding. Clustering of αIIbβ3 also ...
  30. [30]
    Integrin avidity regulation: are changes in affinity and conformation ...
    Central to their function is their unique ability to modulate dynamically their adhesiveness through both affinity- and valency-based mechanisms.
  31. [31]
    α5β1 integrin-mediated adhesion to fibronectin is required for axis ...
    α5β1 integrin-mediated adhesion to fibronectin is required for axis elongation and somitogenesis in mice. PLoS One. 2011;6(7):e22002.<|separator|>
  32. [32]
    Integrin alpha5beta1 and fibronectin regulate polarized cell ...
    Background: Integrin recognition of fibronectin is required for normal gastrulation including the mediolateral cell intercalation behaviors that drive ...
  33. [33]
    Deletion of beta 1 integrins in mice results in inner cell mass failure ...
    Aug 1, 1995 · Heterozygous mutant mice were normal. Homozygous loss of beta 1 integrin expression was lethal during early postimplantation development.<|separator|>
  34. [34]
    Requirement of vascular integrin alpha v beta 3 for angiogenesis
    Integrin alpha v beta 3 was expressed on blood vessels in human wound granulation tissue but not in normal skin, and it showed a fourfold increase in ...
  35. [35]
    Role of alphavbeta3 integrin in the activation of vascular endothelial ...
    Feb 15, 1999 · Alphavbeta3 integrin participates in the full activation of VEGFR-2 triggered by VEGF-A, which is an important angiogenic inducer in tumors, inflammation and ...
  36. [36]
    Anti-angiogenic cancer therapy based on integrin alphavbeta3 ...
    Integrin alphavbeta3 is highly expressed on activated endothelial cells and tumor cells but is not present in resting endothelial cells and most normal organ ...
  37. [37]
    Reduced granulation tissue and wound strength in the absence of ...
    Previous wound healing studies have failed to define a role for either α1β1 or α2β1 integrin in fibroblast-mediated wound contraction, suggesting the ...
  38. [38]
    The integrin-collagen connection--a glue for tissue repair? - PubMed
    Feb 15, 2016 · The bulk of the current literature suggests that collagen-binding integrins only have a limited role in adult connective tissue homeostasis.
  39. [39]
    Integrins promote axonal regeneration after injury of the nervous ...
    α1β1‐integrin ... Fibroblast‐derived tenascin‐C promotes Schwann cell migration through β1‐integrin dependent pathway during peripheral nerve regeneration.
  40. [40]
    Thrombopoietin/MPL signaling regulates hematopoietic stem cell ...
    Dec 13, 2007 · Maintenance of hematopoietic stem cells (HSCs) depends on interaction with their niche ... THPO/MPL signaling upregulated beta1-integrin ...Missing: β1 | Show results with:β1
  41. [41]
    MiR-134-mediated β1 integrin expression and function in ... - PubMed
    Integrins are important molecules within this niche that influence those properties through the interactions of HSCs and mesenchymal stem cells (MSCs). Here we ...
  42. [42]
    The role of β3-integrins in tumor angiogenesis: context is everything
    αvβ3-Integrin has received much attention as a potential anti-angiogenic target because it is upregulated in tumor-associated blood vessels. Agents targeting ...
  43. [43]
    Integrin α6β4 Upregulates PTPRZ1 Through UCHL1-Mediated Hif ...
    Oct 31, 2024 · Integrin α6β4 drives triple-negative breast cancer (TNBC) aggressiveness through the transcriptional regulation of key genes.
  44. [44]
    Cilengitide in newly diagnosed glioblastoma: biomarker expression ...
    Mar 22, 2016 · These trials failed to meet their primary endpoints.Immunohistochemistry was used to assess the levels of the target integrins of cilengitide ...
  45. [45]
    Vedolizumab blocks α4β7 integrin-mediated T cell adhesion to ...
    Jun 28, 2022 · In Crohn's disease and ulcerative colitis, the anti-α4β7 integrin antibody vedolizumab has demonstrated efficacy in phase III trials and has ...
  46. [46]
    Efalizumab in the treatment of psoriasis - PubMed
    The humanized antibody efalizumab is currently the only T-cell directed biologic approved for the treatment of moderate-to-severe psoriasis by both American ...
  47. [47]
    Integrin-β1 is required for the renal cystogenesis caused by ciliary ...
    May 1, 2020 · We showed that the renal cystogenesis caused by loss of Ift88 is prevented when integrin-β1 is simultaneously depleted. In parallel, ...
  48. [48]
    Potential of αvβ6 and αvβ1 integrin inhibition for treatment ... - PubMed
    Jul 2, 2024 · Expert opinion: Bexotegrast, a dual αvβ6/αvβ1 integrin inhibitor, has the potential to slow or even halt disease progression in IPF. Indeed, the ...
  49. [49]
    Glanzmann Thrombasthenia 10 Years Later: Progress Made and ...
    Mar 18, 2024 · On activation αIIbβ3 undergoes conformational changes and binds fibrinogen (Fg) and other proteins to join platelets in the aggregate. The ...
  50. [50]
    SARS-CoV-2 uses major endothelial integrin αvβ3 to cause vascular ...
    Jun 23, 2021 · SARS-CoV-2 uses major endothelial integrin αvβ3 to cause vascular dysregulation in-vitro during COVID-19. PLoS One. 2021 Jun 23;16(6): ...<|separator|>
  51. [51]
    Integrin α9 depletion promotes β-catenin degradation to suppress ...
    Nov 15, 2019 · Experimentally, ITGA9 KO significantly reduced TNBC cell cancer stem cell (CSC)-like property, tumor angiogenesis, tumor growth and metastasis ...
  52. [52]
    Integrin ligands at a glance | Journal of Cell Science
    Oct 1, 2006 · Four α subunits containing an αA-domain (α1, α2, α10 and α11) combine with β1 and form a distinct laminin/collagen-binding subfamily. Few other ...
  53. [53]
    Structure and function of the platelet integrin α IIb β 3 - JCI
    The β3 head consists of a βA domain whose fold resembles that of integrin α subunit “I-domains” and contains a metal ion–dependent adhesion site (MIDAS) motif, ...Missing: organization paper<|control11|><|separator|>
  54. [54]
    The integrin αvβ8 mediates epithelial homeostasis through MT1 ...
    The fact that the cytoplasmic domain of β8 is not required for activation of TGF-β1 suggests that αvβ8-mediated activation of TGF-β might be regulated ...
  55. [55]
    The role of integrins in inflammation and angiogenesis - Nature
    Oct 7, 2020 · Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current ...
  56. [56]
    Evolution of the integrin alpha and beta protein families - PubMed
    Two major families of vertebrate alpha integrins originated prior to the divergence of deuterostomes and protostomes.
  57. [57]
    Comparative Studies of Vertebrate Beta Integrin Genes and Proteins
    Comparative ITB amino acid sequences and protein structures and ITB gene locations were examined using data from several vertebrate genome projects.
  58. [58]
    Functional and expression analysis reveals the involvement of ...
    Aug 19, 2022 · In this study, an integrin αvβ3 (GCαvβ3) originally assembled by αv (GCαv) and β3 (GCβ3) subunits, was identified from a teleost fish grass carp ...
  59. [59]
    Drosophila integrin adhesion complexes are essential for hemocyte ...
    In Drosophila, the integrin family consists of five αPS subunits (αPS1 to 5) and two β subunits, βν and βPS, the latter also known as myospheroid (Brown et al., ...Summary · Introduction · Results · Discussion
  60. [60]
    Analyses of PS integrin functions during Drosophila development
    The common beta subunit of PS integrins (PS beta) is encoded by the lethal(l)myospheroid gene (mys) and is required during wing, eye and muscle development.
  61. [61]
    Roles for βpat-3 Integrins in Development and Function ...
    Unlike vertebrates, which have at least nine β subunit genes, the nematode Caenorhabditis elegans expresses only one β subunit (βpat-3), and a null mutation in ...
  62. [62]
    Cell-cell adhesion via the ECM: integrin genetics in fly and worm
    Just two integrins are present in C. elegans: one putative RGD binding integrin alphapat-2betapat-3, corresponding to Drosophila alphaPS2betaPS and vertebrate ...
  63. [63]
    Invertebrate Integrins: Structure, Function, and Evolution
    The structure of all of the invertebrate subunits is remarkably similar to that of vertebrate integrin subunits. Some experimental data and patterns of ...Missing: non- | Show results with:non-
  64. [64]
    Differences in Regulation of Drosophila and Vertebrate Integrin ...
    In worms, flies, and vertebrates, talin forms a physical link between integrin cytoplasmic domains and the actin cytoskeleton. Loss of either integrins or talin ...Missing: non- | Show results with:non-
  65. [65]
    α integrin cytoplasmic tails have tissue-specific roles during C ...
    Our work in C. elegans established a system of α integrin regulation to control cell migration. In contrast to humans, with 24 different αβ integrin ...
  66. [66]
    Ancient origin of the integrin-mediated adhesion and signaling ...
    The integrin adhesion complex mediates critical interactions between cells and the extracellular matrix, modulating several aspects of cell physiology.Missing: invertebrates mammals
  67. [67]
    Origin of animal multicellularity: precursors, causes, consequences ...
    Feb 5, 2017 · Integrins and associated molecules used for epithelial cell adhesion to extracellular matrix (ECM) were secondarily lost by choanoflagellates ...