Are transmembrane proteins that regulate cellular processes such as cell proliferation and differentiation the extracellular domains of?

Abstract

Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed’s Medical Subject Heading (MeSH) thesaurus.

Impact

  • Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis.

  • This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants.

  • This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.

Introduction

Integrins are a family of ubiquitous αβ heterodimeric receptors that exist in multiple conformations and interact with a diverse group of ligands. These molecules mediate interactions between cells and of these cells with the extracellular matrix (ECM) and thereby serve a critical role in signaling and homeostasis. By facilitating dynamic linkages between the intracellular actin cytoskeleton and the ECM, integrins also transduce both external and internal mechanochemical cues and bi-directional signaling across the plasma membrane.1,2 Integrins are involved in a diverse range of body processes, including cellular survival, inflammation, immunity, infection, thrombosis, angiogenesis, and malignancy. In this review, we highlight the structure and function of integrins; the mechanisms involved in integrin activation and signaling; their role in inflammation, infection, and angiogenesis; and discuss current advances in integrin-targeted therapies. Understanding the factors that regulate integrin structure, function, and signaling would enable us to identify new therapeutic targets.

Structure of integrins

In mammals, the family of integrins is comprised of 24 αβ pairs of heterodimeric transmembrane adhesion receptors and cell-surface proteins. These pairings are known to involve 18 α and 8 β subunits (Fig. 1),3 and their non-covalent associations involve an α and another β subunit (Fig. 2).4 The αβ pairings of integrin subunits dictate the specificity of the integrin to a particular ligand, modulate formation of intracellular adhesion complexes, and regulate downstream signaling.1 Six α (α1–6) and seven β (β1–7) subunits are known to form several unique αβ subunit associations (Fig. 1). Interestingly, the earliest discovered integrins, lymphocyte function-associated antigen 1 (integrin αLβ2) and macrophage antigen 1 (integrin αMβ2), derive their specificity from specific α subunits, but these share the same β subunit.5

Fig. 1: Classification of integrin family.

Are transmembrane proteins that regulate cellular processes such as cell proliferation and differentiation the extracellular domains of?

Integrin heterodimers consists of numerous combinations of α and β subunits. With respect to ligand specificity, integrins are generally classified as collagen-binding integrins (α1β1, α2β1, α10β1, and α11β1), RGD-recognizing integrins (α5β1, αVβ1, αVβ3, αVβ5, αVβ6, αVβ8, and αIIbβ3), laminin-binding integrins (α3β1, α6β1, α7β1, and α6β4), and leukocyte integrins (αLβ2, αMβ2, αXβ2, and αDβ2). The β2 integrin subunit (CD18) can pair with one of the four α subunits (αL-CD11a, αM-CD11b, αX-CD11c, and αD-CD11d), forming leukocyte function-associated antigen-1, Mac1/CR3 (macrophage-1 antigen, complement receptor 3), 150.95/CR4 (complement receptor 4), and CD18/CD11d, respectively. CD11a/CD18 is expressed mainly on all leukocytes, while CD11b/CD18, CD11c/CD18, and CD11d/CD18 are expressed on myeloid cells.106,107 The αMβ2 integrin (also known as CR3, CD11b/CD18, or Mac-1) is found on phagocytic cells and implicated in the adhesion of leucocytes to endothelium and opsonization of microbes. Ligands for CR3 include the complement component iC3b, the intercellular adhesion molecule (1CAM-1), and coagulation factors like fibrinogen and factor X.

Full size image

Fig. 2: Schematic of integrin structure and activation.

Are transmembrane proteins that regulate cellular processes such as cell proliferation and differentiation the extracellular domains of?

Structurally, the αβ integrin subunits are type 1 transmembrane proteins. Each subunit consists of one large multi-domain extracellular segment, one transmembrane helix, and a short cytoplasmic tail. The extracellular region interacts with ECM ligands and is composed of about 1104 (700–1100) residues in the α subunit and 778 residues in the β subunits32 and shorter cytoplasmic domains with 30–50 residues.108 The short cytoplasmic tails are composed of 20–70 amino acids and mediate interactions with intracellular cytoskeletal and signaling proteins.1 In response to intracellular or extracellular stimuli, integrin activation occurs by ligand binding or by the changes on the cytoplasmic domains, resulting in elongation and separation of the legs. Integrins appear in a closed or “bent” conformation on resting cells and display a low binding affinity for ligand rendering them inactive to ligand binding or signal transduction; while once activated, the integrin shape extends to an open conformation leading to a high affinity.109 In a closed conformation, integrins show low ligand-binding affinity, partly due to the bend in the center of the α and β subunits, which brings the ligand-binding site within 5 nm of the cell surface.110 However, when the conformation is open, the two subunits straighten with increased integrin affinity for the ligand.111 The initial binding of extracellular ligand effects separation of the cytoplasmic domains, allowing interaction with signal transduction and cytoskeletal molecules during outside-in signaling, while separation of the cytoplasmic domains by talin and other activators activates the head to enable ligand binding during inside-out signaling.4

Full size image

Integrin α subunit family

The integrin α subunits carry a 200 amino acid “inserted” domain, the I-domain (αI). When present on an integrin, the αI domain is an exclusive ligand-binding site. αI integrins have 13 extracellular domains in 2 subunits, which interact with a variety of ligands. The I-domains are seen in 6 out of the 15 integrin α subunits.

Integrin β subunit family

In humans, integrin β subunits have a cytoplasmic tail that have <75 amino acids in length, except the β4 tail that is about 1000 amino acids long (includes four fibronectin type III repeats).3 The integrin β tails have one or two NPxY/F motifs (x refers to any amino acid) that recognize protein modules, phosphotyrosine-binding domains, that are involved in several signaling and cytoskeletal proteins at the cytoplasmic face of the plasma membrane through phosphorylation of the tyrosine (Y) in the NPxY/F motif.3 The integrin β subunit family includes β1–7, which bind the α subunits in different combinations. The most frequently seen β subunit integrin heterodimers is β1. Although β2 integrins show functional overlap, the corresponding α subunit defines its individual functional properties.6 The β2/CD18 chain has also received attention because of its involvement in several inflammatory receptors such as CD11a/CD18, αLβ2, lymphocyte function-associated antigen-1 (LFA-1); CD11b/CD18, αMβ2, Mac-1, complement receptor 3 (CR3); CD11c/CD18 (αXβ2, p150.95, CR4); and CD11d/CD18, αDβ2; Fig. 1). In these β2 integrins, the α subunits bind specific ligands such as the intercellular adhesion molecules (ICAMs). The non-I-domain α subunits in other integrins, such as the laminin-binding α3, α6, and α7, and others that recognize the arginine (R), glycine (G), aspartic acid (D) (RGD) motif (αV, α8, α5, and αIIb), are also closely related to each other.7 The α subunit of each integrin is the primary determinant of its extracellular ligand specificity. The β chain binds acidic residues in ICAMs and in cytoplasmic adapters such as paxillin, talins, and kindlins to facilitate cellular adhesions with the ECM. Integrins interact with the actin cytoskeleton through the talin- and kindlin-binding motifs present in the cytoplasmic domains of their β subunits.8

Characteristics of specific integrin heterodimers

Integrin αβ heterodimers are divided into four classes (leukocyte, collagen-binding, Arg-Gly-Asp (RGD)-binding, and laminin-binding integrins; Fig. 1) based on evolutionary associations, ligand specificity, and restricted expression on white blood cells (β2 and β7 integrins). Leucocyte integrins have a common β2 chain that is linked to CD-18 and bind receptors such as ICAM and plasma proteins such as complement components C3b and C4b.9 Collagen-binding integrins have a common β1 chain that binds various α chains in integrins α1β1, α2β1, α10β1, and α11β1. The α2β1 integrin binds its primary ligand, collagen,10 and chondroadherin, a matrix protein.11 The RGD-binding integrins have a common αV chain or β1 chain. The RGD peptide motif was first discovered in fibronectin12 but was later found in several other ECM proteins, such as fibronectin,9 osteopontin,13 vitronectin,14 von Willebrand factor (VWF),15 and laminin.16 Among the 24 human integrin subtypes known to date, eight integrin dimers recognize the tripeptide RGD motif within ECM proteins, namely: αVβ1, αVβ3, αVβ5, αVβ6, αVβ8, α5β1, α8β1, and αIIbβ3. Laminin-binding integrins (α3β1, α6β1, α7β1, and α6β4) mediate the adhesion of cells to basement membranes in various tissues.9 The α4β1, α9β1, and α4β7 integrin family binds fibronectin in a RGD-independent manner.9

Integrin–ligand binding and consequent activation

The structure and function of integrins are complex. Integrins bind numerous extracellular ligands, intracellular signaling molecules, and the cytoskeleton in a bivalent-cation-dependent manner with varying specificities. Integrins also have many states with multiple conformations and affinities.

Mechanism of integrin ligand binding and conformational states

Integrins bind cell-surface ligands to promote cellular interactions with the ECM and with other cells in the transduction of complex signals that modulate many cellular processes, such as adhesion, migration, and differentiation. These soluble, ECM, or cell surface-bound ligands may include growth factors, structural constituents of the ECM, proteases, cytokines, plasma proteins, microbial pathogens, or receptors specific to immune cells. The affinity and avidity of a ligand may change actively by inside-out signaling in specific pathways. Ligand affinity may vary with the strength of interaction and dissociation of a monovalent protein and its ligand, where ligand avidity refers to its ability to form multiple combinations of bonds.17

Integrins exist primarily in three conformational states: bent–closed (inactive; the predominant state), extended–closed (active; low affinity or intermediate state), and the extended–open (active; high affinity).18 The affinity of integrins to various inhibitory and stimulatory ligands is modulated by bivalent cations, which induce a range of conformational changes in integrins ranging from a folded, inactive, and low-affinity state to a high-affinity conformation (Fig. 2).19 These conformational changes in the extracellular domains of integrins modulate both ligand binding and downstream cellular signaling.

Integrin activation

The activation of integrins increases the affinity of these molecules to extracellular ligands. Integrin tail domains play a critical role in these steps, and any genetic mutations in these parts of integrins can disrupt downstream intracellular signaling.20 Integrin-mediated signaling across cell membranes is typically bi-directional and termed “outside-in” and “inside-out” signaling.20,21 When integrins interact with ECM ligands, a conformational change allows adherence to downstream adaptor molecules in the cell-membrane plane.22 Once clustered, integrins are able to recruit and activate kinases such as Src family kinases, focal adhesion and scaffold molecules such as the adaptor protein p130CRK-associated substrate/breast cancer anti-estrogen resistance 1 (p130CAS/BCAR1).22 These integrin-associated complexes include discrete active and inactive integrin organizations, which can activate unique signaling pathways.23,24

The extracellular domains of integrins are known to undergo a diverse range of conformational changes that alter the ligand-binding domains. In the cytoplasmic tails of integrins, α-helices are seen as heterodimers,25 and the β-strands often bind intracellular proteins, such as talin or filamin.26,27 The cytoplasmic tail may undergo several specific conformational changes to bind a range of other signal transducers.28,29

Integrin bi-directional inside-out and outside-in signaling

Mechanical stress30 and extracellular chemicals31 can induce rapid conformational changes to cause inside-out activation of integrins.32 Integrins display bi-directional signaling across the plasma membrane. Ligand binding induces extracellular-to-cytoplasm signal transduction, and inside-out signaling or priming regulates integrin-ligand binding conformations (Fig. 2). During integrin activation and signaling, the cytoplasmic tail acts as both a receptor and transmitter of signals. Specifically, during inside-out signaling, the activating signals make an impression on the cytoplasmic tail to induce large conformational changes to the extracellular domain, thereby transforming the integrin from a resting to an active state.33 During outside-in signaling, the binding of a ligand to the extracellular domain of active integrin transmits a conformational change to the cytoplasmic tail, which leads to the activation of kinases and adaptor molecules in the cytosol.1 In contrast, talin and kindlin interaction with the β-cytoplasmic tail can trigger inside-out signaling, leading to integrin activation, clustering, and recruitment of intercellular adaptor proteins to strengthen cellular adhesion. Talin is a large dimeric actin-binding protein and a major regulator of integrin activation, and the regulation of talin–integrin interactions is important in the control of integrin activation and signaling pathways.33 Direct interactions between the talin head and the short cytoplasmic tails of β integrin subunits disrupt inhibitory interactions between α and β integrin subunits.33 This leads to conformational changes in the integrin extracellular domains and consequent increase in their ligand affinity. The role of kindlins are not clearly defined, but they are structurally related to the talin head. The synergistic binding of talin and kindlin to β integrin cytoplasmic tails induces integrin activation by disrupting the α–β interactions at the transmembrane and the cytoplasmic domains.33,34

Integrins in inflammation and infection

In the resting state, β2 integrins are expressed specifically on leucocyte receptors. During inflammation, the inflammatory cytokines activate these integrins and promote cellular adherence to the counter-receptors such as ICAMs and promote phagocytosis and cytotoxic killing. Integrin receptors on leukocytes, such as the macrophage-1 antigen (Mac-1, also known as CR3, αMβ2, CD11b/CD18) interact with platelet antigens such as the glycoprotein Ibα (GPIbα) during inflammation. Integrins bind to the pro-domain of transforming growth factor (TGF)-β1 to activate it and promote its secretion. The pro-TGF-βs are biosynthesized and stored in tissues in latent forms, and integrins αVβ6 and αVβ8 can uniquely bind and activate pro-TGF-β1 and pro-TGF-β3. The αVβ6 integrin is known to specifically bind the RGDLXXL/I motif in TGF-β1 and TGF-β3.35

β2 integrins promote recruitment of leukocytes to the sites of inflammation by promoting the adhesion of circulating leukocytes to vascular endothelium, transendothelial migration,36,37 the formation of immunological synapses in leucocytes,38 and inflammatory signaling in involved cells.39 Activated β2 integrins on dendritic cells (DCs) may act as negative regulators of DC migration in certain conditions and may also regulate T cell activation.40,41 β integrins on the leukocyte surface are also involved in the tethering, rolling, and adhesion of leukocytes to activated endothelial cells.42 β2 integrins can also initiate intracellular signaling pathways in macrophages and neutrophils and stimulate cytokine secretion from these cells either directly or in synergy with Toll-like receptors (TLRs).43 Integrins may also integrate the impact of the epidermal growth factor receptor, platelet-derived growth factor receptor, insulin receptor, met receptor superfamily (hepatocyte growth factor receptor), and the vascular endothelial growth factor receptor (VEGFR) in inflammatory cells.44

β2 integrins are important regulators of adhesion, leukocyte recruitment, and immunological signaling. These integrins mediate adhesive interactions between myeloid cells, endothelial cells, antigen-presenting cells, T cells and the ECM.45 L-selectin, the CCR7 chemokine receptor, interacts with specific carbohydrate epitopes on the endothelium and promotes leukocyte rolling and transmigration through the vascular endothelium.46 Leukocyte rolling induces a rapid, although transient, increase in the affinity of the β1 and β2 integrins to the endothelial ligands.47,48 Conformational changes in the structure of the inserted (I) domain of the αL subunit of LFA-149 enhance firm leukocyte adhesion under shear flow.31,49

Role of integrins in neonatal organs during normal development and inflammation

Integrins in the lung during normal development and in inflammation

Integrins and receptor tyrosine kinases act with cytokine and growth factors to modulate the extracellular signal-regulated kinase and phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathways during regeneration, inflammation, developmental, and pathological processes in the developing lung.2,44,50,51 The ECM in the lung contains collagen, fibronectin, laminin, and entactin,52 and alterations in the formation and structure of the ECM during normal development, healing from injury, or in chronic lung disease could lead to profound alterations in the lung structure.53,54 For instance, fibronectin in the ECM promotes integrin-mediated cellular migration and differentiation of cells during lung development.55 β1 integrin activates several signaling pathways, particularly the PI3K/AKT pathway activated during wound healing in the presence of collage VI in the lung ECM.56 β1 integrins play a critical role in alveolar homeostasis, as seen in chronic lung disease depicted in β1 integrin-deficient mice.57 In addition, β1 integrin-deficient alveolar epithelial cells produce excessive monocyte chemoattractant protein 1 and reactive oxygen species, suggesting that β1 integrins may be involved in alveolar homeostasis.58 In murine models of bronchopulmonary dysplasia, perinatal exposure to lipopolysaccharide and increased expression of interleukin-33 may activate neutrophils and promote fibronectin degradation in alveolar epithelial cells.59 Other studies have noted increased expression of integrin α2β1 on mast cells and activation/release of inflammatory cytokines.60,61 Similar findings have been noted in murine models with Listeria monocytogenes infections.62 Mice deficient in integrin α263 and integrin αIIb64 show defective platelet interaction with collagen. α2β1 integrin-null mice have normal angiogenesis but may have altered angiogenic responses during injury repair.65 In contrast, integrin β1 knockout mice may have altered development and are not viable, indicating an essential role of β1 during development. Table 1 outlines murine models of integrins, their target tissues, and signaling.

Table 1 Integrin-targeted murine models and the effect of their signal modulation.

Full size table

Integrins in intestinal inflammation and in necrotizing enterocolitis (NEC)

The regulation of intestinal leukocyte responses is vital to maintaining immune homeostasis and prevention of intestinal inflammatory conditions. Integrin αvβ5 is expressed on neonatal intestinal macrophages; the expression is developmentally regulated and is not dependent on microbial colonization. These integrins bind different ECM components, such as laminins, collagens, and fibronectin, and are known to coordinate epithelial cell adhesion and movement.4,66 These integrins recognize the RGD tri-peptide sequence present in ECM proteins, such as fibronectin and vitronectin.67,68 The integrin αVβ5 can be found in both focal adhesions and in clathrin-coated membrane domains.69,70

Integrin αvβ8 plays an important role in epithelial homeostasis and is a major activator of TGF-β expression.71 α3 and β1 integrins, which are known to increase epithelial migration, are upregulated by bacterial products during NEC.72,73 TLR4 signaling on enterocytes promotes the efflux of β1 integrins from the cytoplasm toward the cell membrane and enhances cell–matrix contacts that limit cellular movement.74 In other studies, Besner and colleagues have examined the role of E-cadherin and integrins in NEC and showed that the growth factor, heparin-bound epidermal growth factor, can promote intestinal restitution in NEC through its effects on integrin–ECM interaction and intercellular adhesions.75 Intestinal epithelial cells also express α3β1, another set of integrins of translational importance. In NEC, increased epithelial expression of α3β1 may impair the migration of epithelial cells needed for mucosal wound healing.74 However, the same α3 integrins are also required for morphologic differentiation of the intestinal epithelium in the developing intestine.76 Despite the physiological needs of the β1 integrins, therapeutic targeting of these molecules may still be possible with information on the best timing and the possibility of regionally focused intervention.

Integrins in the developing eye and in retinopathy of prematurity

In the developing eye, disruption of the oxygen supply to the retina can disrupt neuronal dysfunction needed for transduction and transmission of photosensitive visual signals to the occipital lobe and other cognitive centers. Integrin α2β1 and VEGF interact closely in several intracellular angiogenic signaling (Fig. 3).77,78 Cyclic peptides selectively inhibit αVβ3 and αVβ5, and are potent inhibitors of endothelial cell invasion and differentiation induced by VEGF-A or fibroblast growth factor-2.78 Integrin αVβ3 works synergistically with VEGF to activate angiogenesis in endothelial cells via VEGFR‐2 phosphorylation.79 Endothelial cells are the primary cells expressing both VEGFR-2 and α2β1 integrin.80 Proteoglycans such as decorin and perlecan in the ECM of the eye can modulate α2β1 and play a vital role in angiogenesis.80 The C-terminal fragment of perlecan, known as endorepellin, has an opposite effect and blocks angiogenesis through antagonism of VEGFR-2 and α2β1 integrin on endothelial cells.80 Retinal pigment epithelial cells express beta‐8 integrin at the surface, and the knockdown of beta-8 integrin significantly decreased retinal pigment epithelial cell migration in wound-healing assays.81

Fig. 3: Schematic of integrin regulation of angiogenic signaling.

Are transmembrane proteins that regulate cellular processes such as cell proliferation and differentiation the extracellular domains of?

The schematic shows the interaction between the signaling pathways regulated by αβ integrins and the VEGF receptor. VEGF-A promotes angiogenesis through VEGF receptor-2 (VEGFR2), a tyrosine kinase receptor expressed by endothelial cells.112 When VEGF-A binds to VEGFR2, numerous intracellular signaling pathways are activated, such as phosphatidylinositol 3-kinase (PI3K), extracellular signal-regulated kinase (Erk), focal adhesion kinase (FAK), c-Src family, and paxillin, a signal transduction adaptor protein associated with focal adhesion.113,114 Specifically, FAK phosphorylates its substrate, paxillin, which activates ERK signaling.114 When integrins activate the tyrosine phosphorylation of FAK, it binds to signaling structural proteins, PI3K, and paxillin.115 Src family kinases (SFKs) play a critical role in cell adhesion, survival, and angiogenesis, interact with VEGF receptor, regulate gene expression of angiogenic growth factors, modulate cell proliferation via the mitogen-activated protein kinases (MAPK)-ERK pathway, and interact with integrins to regulate cell adhesion and migration. ECM extracellular matrix, VEGF vascular endothelial growth factor.

Full size image

The retinal tissue has one of the body’s highest metabolic demands, placing it at risk of injury from oxidative stress, metabolic derangements, and consequent pathologic neovascularization seen in retinopathy of prematurity (ROP) and other proliferative retinal vitreoretinopathies. ROP is a bi-phasic disease of retinal vascular development due to dysregulation of VEGF.82,83 In phase 1, VEGF is downregulated during exposure to hyperoxia, while in phase 2, VEGF is upregulated in relative/true hypoxia. VEGF is known to have several isoforms; VEGFA165 is the predominant isoform in the eye with multiple pro- and anti-angiogenic splice variants.84 In a newborn mouse model of oxygen-induced retinopathy (OIR), oxidative stress from fluctuating hyperoxia and hypoxia leads to altered vascular development with tortuous arteries, dilated veins, and capillary attrition, akin to human ROP.82,85 These changes persist in adult mice with long-term abnormalities in vascularization, structure, and function both in vivo and histologically.86,87

Integrin-targeted therapy holds promise in ROP. Targeting α2β1 integrin expression on endothelial cells mitigates OIR,88 and the administration of 3-[3-(6-guanidino-1-oxoisoindolin-2-yl) propanamido]-3-(pyridin-3-yl) propanoic acid dihydrochloride, a novel non-peptide αvβ3 antagonist, can inhibit retinal neovascularization.89 There are exciting possibilities that endothelial α2β1 may be therapeutic target in pathological angiogenesis.

Integrins in thrombosis and fibrosis

Platelet adhesion and signaling play key roles in hemostasis and thrombosis. Two platelet receptors, integrin αIIbβ3 and GPIbα, mediate the early and mid-stages of platelet adhesion in the vascular environment.90 GPIbα is a key part of the receptor for VWF, and its binding to VWF enables platelet rolling during the formation of thrombotic plugs at the sites of vascular injury.91,92,93 αIIbβ3 is expressed on both platelets and the endothelium, and upon activation, it promotes platelet adhesion and aggregation by cross-linking with soluble fibrinogen, fibronectin, and VWF.

In alloimmune thrombocytopenia, autoantibodies are frequently seen against integrin β3 and GPIbα.94,95 Intracranial hemorrhages may be seen more frequently in infants with anti-β3 integrin antibodies than in those with antibodies against GPIbα.96 Existing in vitro and in vivo data suggest that the β3 integrin may bind a wider range of ligands, including fibrinogen and VWF, and autoantibodies that block its function may induce a deeper functional deficit than the anti-GPIbα antibodies.97

Integrin-targeted therapies

Integrin dysregulation is implicated in the pathogenesis of numerous diseases with altered angiogenesis, inflammation, or in infectious diseases. In these conditions, therapeutic strategies may either directly target integrins or their ligands. Out of the 24 known human integrins, many have already been identified as therapeutic targets for monoclonal antibodies, peptides, and/or small molecules. In adult subjects, efforts are ongoing to target platelet integrin αIIbβ3 to prevent thrombotic complications after percutaneous vascular interventions, lymphocyte α4β1 and α4β7 integrins in the treatment of multiple sclerosis, and β7 integrins (α4β7 and αEβ7 integrins) in inflammatory bowel disease.98 Specifically, a humanized anti-α4 antibody (Natalizumab) works in reduction of inflammation in multiple sclerosis by blocking the α4β1–vascular cell adhesion molecule interaction or the α4β7–mucosal addressin cell adhesion molecule interaction on mucosal endothelium and blocking leukocyte trafficking across the blood–brain barrier.99 In another study, a micellar delivery vehicle decorated with an anti-angiogenic peptide has been shown to inhibit αVβ3-mediated neovascularization in endothelial cells.100 Several anticancer drugs have also been developed against integrin ligands or by using integrin-targeted encapsulated nanoparticles as vehicles to unload drugs into the vasculature of several tumors.101 In a mouse model of hepatic fibrosis, cyclic peptide-guided liposomes preferentially targeted the activated hepatic stellate cells (not quiescent ones) to treat the fibrotic phenotype.102 αVβ3 antagonists are being tried for the inhibition of retinal neovascularization and may have therapeutic value in ROP.89 In a mouse model of laser-induced choroidal neovascularization, intravenous injection of irradiated nanoparticles loaded with doxorubicin allowed nanoparticle accumulation in the neovascular lesions and reduced the size of neovascular lesions.103 Integrin antagonists may also be used in fibrotic diseases; IDL-2965 is being studied as a selective, highly potent, anti-fibrotic integrin antagonist in idiopathic pulmonary fibrosis.104 Small molecule pure antagonists, TDI-4161 and TDI-3761, have been designed to inhibit αVβ3-mediated cell adhesion to αVβ3 ligands.105 Further studies are needed to improve the specificity of anti-integrin drugs to improve both the safety profile and therapeutic success of these agents.

Conclusions

Enhanced understanding of integrin ligand interactions will enable development of therapies targeting specific receptors in order to modulate angiogenic, thrombotic, infections, and inflammatory disorders. Although numerous animal studies have shown promise in the clinical use of integrins as therapeutic targets, there is a need for clinical studies to confirm efficacy and safety in neonates and young infants. In this review, we have summarized and outlined the roles of integrins in inflammation, angiogenesis, and infectious conditions. Therapies could be targeted specifically to alpha subunits, but their overlapping roles are a critical factor to be considered. Further studies are needed both on molecular signaling and regulatory mechanisms of integrin function and the safety and efficacy in clinical settings.

References

  1. Morse, E. M., Brahme, N. N. & Calderwood, D. A. Integrin cytoplasmic tail interactions. Biochemistry 53, 810–820 (2014).

    CAS  PubMed  Google Scholar 

  2. Cooper, J. & Giancotti, F. G. Integrin signaling in cancer: mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell 35, 347–367 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  3. Takada, Y., Ye, X. & Simon, S. The integrins. Genome Biol. 8, 215 (2007).

    PubMed Central  PubMed  Google Scholar 

  4. Hynes, R. O. Integrins: bidirectional, allosteric signaling machines. Cell 110, 673–687 (2002).

    CAS  PubMed  Google Scholar 

  5. Kurzinger, K., Ho, M. K. & Springer, T. A. Structural homology of a macrophage differentiation antigen and an antigen involved in T-cell-mediated killing. Nature 296, 668–670 (1982).

    CAS  PubMed  Google Scholar 

  6. Bednarczyk, M., Stege, H., Grabbe, S. & Bros, M. β2 Integrins—multi-functional leukocyte receptors in health and disease. Int. J. Mol. Sci. 21, 1402 (2020).

    CAS  PubMed Central  Google Scholar 

  7. Lee, J. O., Bankston, L. A., Arnaout, M. A. & Liddington, R. C. Two conformations of the integrin A-domain (I-domain): a pathway for activation? Structure 3, 1333–1340 (1995).

    CAS  PubMed  Google Scholar 

  8. McCarty, J. H., Cook, A. A. & Hynes, R. O. An interaction between αvβ8 integrin and Band 4.1B via a highly conserved region of the band 4.1 C-terminal domain. Proc. Natl Acad. Sci. USA 102, 13479–13483 (2005).

    CAS  PubMed Central  PubMed  Google Scholar 

  9. Koivisto, L., Heino, J., Häkkinen, L. & Larjava, H. Integrins in wound healing. Adv. Wound Care 3, 762–783 (2014).

    Google Scholar 

  10. Emsley, J., Knight, C. G., Farndale, R. W., Barnes, M. J. & Liddington, R. C. Structural basis of collagen recognition by integrin alpha2beta1. Cell 101, 47–56 (2000).

    CAS  PubMed  Google Scholar 

  11. Haglund, L. et al. Identification and characterization of the integrin alpha2beta1 binding motif in chondroadherin mediating cell attachment. J. Biol. Chem. 286, 3925–3934 (2011).

    CAS  PubMed  Google Scholar 

  12. Pierschbacher, M. D. & Ruoslahti, E. Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature 309, 30–33 (1984).

    CAS  PubMed  Google Scholar 

  13. Oldberg, A., Franzen, A. & Heinegard, D. Cloning and sequence analysis of rat bone sialoprotein (osteopontin) cDNA reveals an Arg-Gly-Asp cell-binding sequence. Proc. Natl Acad. Sci. USA 83, 8819–8823 (1986).

    CAS  PubMed Central  PubMed  Google Scholar 

  14. Suzuki, S., Oldberg, A., Hayman, E. G., Pierschbacher, M. D. & Ruoslahti, E. Complete amino acid sequence of human vitronectin deduced from cDNA. Similarity of cell attachment sites in vitronectin and fibronectin. EMBO J. 4, 2519–2524 (1985).

    CAS  PubMed Central  PubMed  Google Scholar 

  15. Plow, E. F., Pierschbacher, M. D., Ruoslahti, E., Marguerie, G. A. & Ginsberg, M. H. The effect of Arg-Gly-Asp-containing peptides on fibrinogen and von Willebrand factor binding to platelets. Proc. Natl Acad. Sci. USA 82, 8057–8061 (1985).

    CAS  PubMed Central  PubMed  Google Scholar 

  16. Grant, D. S. et al. Two different laminin domains mediate the differentiation of human endothelial cells into capillary-like structures in vitro. Cell 58, 933–943 (1989).

    CAS  PubMed  Google Scholar 

  17. Abram, C. L. & Lowell, C. A. The ins and outs of leukocyte integrin signaling. Annu. Rev. Immunol. 27, 339–362 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  18. Nishida, N. et al. Activation of leukocyte β2 integrins by conversion from bent to extended conformations. Immunity 25, 583–594 (2006).

    CAS  PubMed  Google Scholar 

  19. Mould, A. P., Garratt, A. N., Puzon-McLaughlin, W., Takada, Y. & Humphries, M. J. Regulation of integrin function: evidence that bivalent-cation-induced conformational changes lead to the unmasking of ligand-binding sites within integrin alpha5 beta1. Biochem. J. 331(Pt 3), 821–828 (1998).

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Calderwood, D. A. Integrin activation. J. Cell Sci. 117, 657–666 (2004).

    CAS  PubMed  Google Scholar 

  21. Van Agthoven, J. F. et al. Structural basis for pure antagonism of integrin alphaVbeta3 by a high-affinity form of fibronectin. Nat. Struct. Mol. Biol. 21, 383–388 (2014).

    PubMed Central  PubMed  Google Scholar 

  22. Desgrosellier, J. S. & Cheresh, D. A. Integrins in cancer: biological implications and therapeutic opportunities. Nat. Rev. Cancer 10, 9 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Li, J. & Springer, T. A. Integrin extension enables ultrasensitive regulation by cytoskeletal force. Proc. Natl Acad. Sci. USA 114, 4685–4690 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  24. De Mets, R. et al. Cellular tension encodes local Src-dependent differential beta1 and beta3 integrin mobility. Mol. Biol. Cell 30, 181–190 (2019).

    PubMed Central  PubMed  Google Scholar 

  25. Bhunia, A., Tang, X. Y., Mohanram, H., Tan, S. M. & Bhattacharjya, S. NMR solution conformations and interactions of integrin alphaLbeta2 cytoplasmic tails. J. Biol. Chem. 284, 3873–3884 (2009).

    PubMed  Google Scholar 

  26. Kiema, T. et al. The molecular basis of filamin binding to integrins and competition with talin. Mol. Cell 21, 337–347 (2006).

    CAS  PubMed  Google Scholar 

  27. Ellis, S. J. et al. The talin head domain reinforces integrin-mediated adhesion by promoting adhesion complex stability and clustering. PLoS Genet. 10, e1004756 (2014).

    PubMed Central  PubMed  Google Scholar 

  28. Beglova, N., Blacklow, S. C., Takagi, J. & Springer, T. A. Cysteine-rich module structure reveals a fulcrum for integrin rearrangement upon activation. Nat. Struct. Biol. 9, 282–287 (2002).

    CAS  PubMed  Google Scholar 

  29. Armulik, A., Nilsson, I., von Heijne, G. & Johansson, S. Determination of the border between the transmembrane and cytoplasmic domains of human integrin subunits. J. Biol. Chem. 274, 37030–37034 (1999).

    CAS  PubMed  Google Scholar 

  30. Zwartz, G. J. et al. Real-time analysis of very late antigen-4 affinity modulation by shear. J. Biol. Chem. 279, 38277–38286 (2004).

    CAS  PubMed  Google Scholar 

  31. Constantin, G. et al. Chemokines trigger immediate β2 integrin affinity and mobility changes: differential regulation and roles in lymphocyte arrest under flow. Immunity 13, 759–769 (2000).

    CAS  PubMed  Google Scholar 

  32. Arnaout, M. A., Mahalingam, B. & Xiong, J.-P. Integrin structure, allostery, and bidirectional signaling. Annu. Rev. Cell Dev. Biol. 21, 381–410 (2005).

    CAS  PubMed  Google Scholar 

  33. Calderwood, D. A., Campbell, I. D. & Critchley, D. R. Talins and kindlins: partners in integrin-mediated adhesion. Nat. Rev. Mol. Cell Biol. 14, 503–517 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Kim, C., Ye, F. & Ginsberg, M. H. Regulation of integrin activation. Annu. Rev. Cell Dev. Biol. 27, 321–345 (2011).

    CAS  PubMed  Google Scholar 

  35. Dong, X., Hudson, N. E., Lu, C. & Springer, T. A. Structural determinants of integrin beta-subunit specificity for latent TGF-beta. Nat. Struct. Mol. Biol. 21, 1091–1096 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  36. von Andrian, U. H. et al. Two-step model of leukocyte-endothelial cell interaction in inflammation: distinct roles for LECAM-1 and the leukocyte beta 2 integrins in vivo. Proc. Natl Acad. Sci. USA 88, 7538–7542 (1991).

    Google Scholar 

  37. Lammermann, T. et al. Rapid leukocyte migration by integrin-independent flowing and squeezing. Nature 453, 51–55 (2008).

    PubMed  Google Scholar 

  38. Monks, C. R., Freiberg, B. A., Kupfer, H., Sciaky, N. & Kupfer, A. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature 395, 82–86 (1998).

    CAS  PubMed  Google Scholar 

  39. Szukiewicz, D. et al. Chorioamnionitis (ChA) modifies CX3CL1 (fractalkine) production by human amniotic epithelial cells (HAEC) under normoxic and hypoxic conditions. J. Inflamm. 11, 12 (2014).

    Google Scholar 

  40. Varga, G. et al. Active MAC-1 (CD11b/CD18) on DCs inhibits full T-cell activation. Blood 109, 661–669 (2007).

    CAS  PubMed  Google Scholar 

  41. Schittenhelm, L., Hilkens, C. M. & Morrison, V. L. β2 integrins as regulators of dendritic cell, monocyte, and macrophage function. Front. Immunol. 8, 1866 (2017).

    PubMed Central  PubMed  Google Scholar 

  42. Herter, J. & Zarbock, A. Integrin regulation during leukocyte recruitment. J. Immunol. 190, 4451–4457 (2013).

    CAS  PubMed  Google Scholar 

  43. Wolf, D. et al. A ligand-specific blockade of the integrin Mac-1 selectively targets pathologic inflammation while maintaining protective host-defense. Nat. Commun. 9, 1–11 (2018).

    CAS  Google Scholar 

  44. Giancotti, F. G. & Tarone, G. Positional control of cell fate through joint integrin/receptor protein kinase signaling. Annu. Rev. Cell Dev. Biol. 19, 173–206 (2003).

    CAS  PubMed  Google Scholar 

  45. Shimizu, Y., Rose, D. M. & Ginsberg, M. H. Integrins in the immune system. Adv. Immunol. 72, 325–380 (1999).

  46. Lasky, L. A. et al. An endothelial ligand for L-selectin is a novel mucin-like molecule. Cell 69, 927–938 (1992).

    CAS  PubMed  Google Scholar 

  47. Grabovsky, V. et al. Subsecond induction of alpha4 integrin clustering by immobilized chemokines stimulates leukocyte tethering and rolling on endothelial vascular cell adhesion molecule 1 under flow conditions. J. Exp. Med. 192, 495–506 (2000).

    CAS  PubMed Central  PubMed  Google Scholar 

  48. Chen, C. et al. High affinity very late antigen-4 subsets expressed on T cells are mandatory for spontaneous adhesion strengthening but not for rolling on VCAM-1 in shear flow. J. Immunol. 162, 1084–1095 (1999).

    CAS  PubMed  Google Scholar 

  49. Salas, A., Shimaoka, M., Chen, S., Carman, C. V. & Springer, T. Transition from rolling to firm adhesion is regulated by the conformation of the I domain of the integrin lymphocyte function-associated antigen-1. J. Biol. Chem. 277, 50255–50262 (2002).

    CAS  PubMed  Google Scholar 

  50. Danen, E. H. & Yamada, K. M. Fibronectin, integrins, and growth control. J. Cell. Physiol. 189, 1–13 (2001).

    CAS  PubMed  Google Scholar 

  51. Mižíková, I. & Morty, R. E. The extracellular matrix in bronchopulmonary dysplasia: target and source. Front. Med. 2, 91 (2015).

    Google Scholar 

  52. Kahsai, T. Z. et al. Seminiferous tubule basement membrane. Composition and organization of type IV collagen chains, and the linkage of alpha3(IV) and alpha5(IV) chains. J. Biol. Chem. 272, 17023–17032 (1997).

    CAS  PubMed  Google Scholar 

  53. Vlahovic, G., Russell, M. L., Mercer, R. R. & Crapo, J. D. Cellular and connective tissue changes in alveolar septal walls in emphysema. Am. J. Respir. Crit. Care Med. 160, 2086–2092 (1999).

    CAS  PubMed  Google Scholar 

  54. Burgstaller, G. et al. The instructive extracellular matrix of the lung: basic composition and alterations in chronic lung disease. Eur. Respir. J. 50, 1601805 (2017).

    PubMed  Google Scholar 

  55. Roy, D. C. & Hocking, D. C. Recombinant fibronectin matrix mimetics specify integrin adhesion and extracellular matrix assembly. Tissue Eng. Part A 19, 558–570 (2013).

    CAS  PubMed  Google Scholar 

  56. Mereness, J. A. et al. Type VI collagen promotes lung epithelial cell spreading and wound-closure. PLoS ONE 13, e0209095 (2018).

    PubMed Central  PubMed  Google Scholar 

  57. Plosa, E. J. et al. β1 integrin regulates adult lung alveolar epithelial cell inflammation. JCI Insight 5, e129259 (2020).

  58. Plosa, E. J. et al. Epithelial β1 integrin is required for lung branching morphogenesis and alveolarization. Development 141, 4751–4762 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  59. Jin, R. et al. IL-33-induced neutrophil extracellular traps degrade fibronectin in a murine model of bronchopulmonary dysplasia. Cell Death Discov. 6, 33 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Ringel-Scaia, V. M., Powell, M. D., Read, K. A., Allen, I. C. & Oestreich, K. J. Systemic Listeria monocytogenes infection as a model to study T helper cell immune responses. Methods Mol. Biol. 1960, 149–160 (2019).

  61. McCall-Culbreath, K. D., Li, Z. & Zutter, M. M. Crosstalk between the α2β1 integrin and c-met/HGF-R regulates innate immunity. Blood 111, 3562–3570 (2008).

    CAS  PubMed Central  PubMed  Google Scholar 

  62. Edelson, B. T., Li, Z., Pappan, L. K. & Zutter, M. M. Mast cell–mediated inflammatory responses require the α2β1 integrin. Blood 103, 2214–2220 (2004).

    CAS  PubMed  Google Scholar 

  63. Holtkotter, O. et al. Integrin alpha 2-deficient mice develop normally, are fertile, but display partially defective platelet interaction with collagen. J. Biol. Chem. 277, 10789–10794 (2002).

    CAS  PubMed  Google Scholar 

  64. Bouvard, D. et al. Functional consequences of integrin gene mutations in mice. Circ. Res. 89, 211–223 (2001).

    CAS  PubMed  Google Scholar 

  65. Chen, J., Diacovo, T. G., Grenache, D. G., Santoro, S. A. & Zutter, M. M. The alpha(2) integrin subunit-deficient mouse: a multifaceted phenotype including defects of branching morphogenesis and hemostasis. Am. J. Pathol. 161, 337–344 (2002).

    CAS  PubMed Central  PubMed  Google Scholar 

  66. Barczyk, M., Carracedo, S. & Gullberg, D. Integrins. Cell Tissue Res. 339, 269 (2010).

    CAS  PubMed  Google Scholar 

  67. Bodary, S. & McLean, J. W. The integrin beta 1 subunit associates with the vitronectin receptor alpha v subunit to form a novel vitronectin receptor in a human embryonic kidney cell line. J. Biol. Chem. 265, 5938–5941 (1990).

    CAS  PubMed  Google Scholar 

  68. Charo, I. F., Nannizzi, L., Smith, J. W. & Cheresh, D. A. The vitronectin receptor avb3 binds fibronectin and acts in concert with a5b1 in promoting cellular attachment and spreading on fibronectin. J. Cell Biol. 111, 2795–2800 (1990).

    CAS  PubMed  Google Scholar 

  69. Grove, J. et al. Flat clathrin lattices: stable features of the plasma membrane. Mol. Biol. Cell 25, 3581–3594 (2014).

    PubMed Central  PubMed  Google Scholar 

  70. Lampe, M., Vassilopoulos, S. & Merrifield, C. Clathrin coated pits, plaques and adhesion. J. Struct. Biol. 196, 48–56 (2016).

    CAS  PubMed  Google Scholar 

  71. Sherman, M. P. New concepts of microbial translocation in the neonatal intestine: mechanisms and prevention. Clin. Perinatol. 37, 565–579 (2010).

    PubMed Central  PubMed  Google Scholar 

  72. Neu J., Pammi M. Pathogenesis of NEC: impact of an altered intestinal microbiome. Semin. Perinatol. 41, 29–35 (2017).

  73. Fundora, J. B., Guha, P., Shores, D. R., Pammi, M. & Maheshwari, A. Intestinal dysbiosis and necrotizing enterocolitis: assessment for causality using Bradford Hill criteria. Pediatr. Res. 87, 235–248 (2019).

  74. Qureshi, F. G. et al. Increased expression and function of integrins in enterocytes by endotoxin impairs epithelial restitution. Gastroenterology 128, 1012–1022 (2005).

    CAS  PubMed  Google Scholar 

  75. Su, Y., Yang, J. & Besner, G. E. HB-EGF promotes intestinal restitution by affecting integrin–extracellular matrix interactions and intercellular adhesions. Growth Factors 31, 39–55 (2013).

    CAS  PubMed  Google Scholar 

  76. Zhang, X., Cromwell, J. W., Kunjummen, B. D., Yee, D. & Garcia-Aguilar, J. The alpha2 and alpha3 integrins are required for morphologic differentiation of an intestinal epithelial cell line. Surgery 133, 429–437 (2003).

    PubMed  Google Scholar 

  77. Campochiaro, P. A., Aiello, L. P. & Rosenfeld, P. J. Anti–vascular endothelial growth factor agents in the treatment of retinal disease: from bench to bedside. Ophthalmology 123, S78–S88 (2016).

    PubMed  Google Scholar 

  78. Nisato, R. E., Tille, J.-C., Jonczyk, A., Goodman, S. L. & Pepper, M. S. αvβ3 and αvβ5 integrin antagonists inhibit angiogenesis in vitro. Angiogenesis 6, 105–119 (2003).

    CAS  PubMed  Google Scholar 

  79. Soldi, R. et al. Role of αvβ3 integrin in the activation of vascular endothelial growth factor receptor‐2. EMBO J. 18, 882–892 (1999).

    CAS  PubMed Central  PubMed  Google Scholar 

  80. Douglass, S., Goyal, A. & Iozzo, R. V. The role of perlecan and endorepellin in the control of tumor angiogenesis and endothelial cell autophagy. Connect. Tissue Res. 56, 381–391 (2015).

    PubMed Central  PubMed  Google Scholar 

  81. Caceres, P. S., Hanke-Gogokhia, C., Ridano, M. E., Zobel, G. & Rodriguez-Boulan, E. Cell‐cell and cell‐extracellular matrix communication pathways identified in the polarized surface proteome of retinal pigment epithelial cells. FASEB J. 34, 1–1 (2020).

    Google Scholar 

  82. Mezu-Ndubuisi, O. J. In vivo angiography quantifies oxygen-induced retinopathy vascular recovery. Optom. Vis. Sci. 93, 1268–1279 (2016).

    PubMed Central  PubMed  Google Scholar 

  83. Mezu-Ndubuisi, O. J. et al. Intravitreal delivery of VEGF-A165-loaded PLGA microparticles reduces retinal vaso-obliteration in an in vivo mouse model of retinopathy of prematurity. Curr. Eye Res. 44, 275–286 (2019).

    CAS  PubMed  Google Scholar 

  84. Zhao, M. et al. Expression of pro-and anti-angiogenic isoforms of VEGF in the mouse model of oxygen-induced retinopathy. Exp. Eye Res. 93, 921–926 (2011).

    CAS  PubMed  Google Scholar 

  85. Mezu-Ndubuisi, O. J. et al. In vivo retinal vascular oxygen tension imaging and fluorescein angiography in the mouse model of oxygen-induced retinopathy. Investig. Ophthalmol. Vis. Sci. 54, 6968–6972 (2013).

    Google Scholar 

  86. Mezu-Ndubuisi, O. J., Adams, T., Taylor, L. K., Nwaba, A. & Eickhoff, J. Simultaneous assessment of aberrant retinal vascularization, thickness, and function in an in vivo mouse oxygen-induced retinopathy model. Eye 33, 363–373 (2019).

    PubMed  Google Scholar 

  87. Mezu-Ndubuisi, O. J. et al. Long-term evaluation of retinal morphology and function in a mouse model of oxygen-induced retinopathy. Mol. Vis. 26, 257–276 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  88. Madamanchi, A. et al. Mitigation of oxygen-induced retinopathy in α2β1 integrin-deficient mice. Investig. Ophthalmol. Vis. Sci. 55, 4338–4347 (2014).

    CAS  Google Scholar 

  89. Li, Y.-J. et al. Therapeutic efficacy of a novel non-peptide αvβ3 integrin antagonist for pathological retinal angiogenesis in mice. Exp. Eye Res. 129, 119–126 (2014).

    CAS  PubMed  Google Scholar 

  90. Monti, M. et al. Integrin-dependent cell adhesion to neutrophil extracellular traps through engagement of fibronectin in neutrophil-like cells. PLoS ONE 12, e0171362 (2017).

    PubMed Central  PubMed  Google Scholar 

  91. Ruggeri, Z. M. Mechanisms initiating platelet thrombus formation. Thrombosis Haemost. 78, 611–616 (1997).

    CAS  Google Scholar 

  92. Savage, B., Almus-Jacobs, F. & Ruggeri, Z. M. Specific synergy of multiple substrate–receptor interactions in platelet thrombus formation under flow. Cell 94, 657–666 (1998).

    CAS  PubMed  Google Scholar 

  93. Tsuji, S. et al. Real-time analysis of mural thrombus formation in various platelet aggregation disorders: distinct shear-dependent roles of platelet receptors and adhesive proteins under flow. Blood. J. Am. Soc. Hematol. 94, 968–975 (1999).

    CAS  Google Scholar 

  94. Brooks, P. C., Clark, R. A. & Cheresh, D. A. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 264, 569–571 (1994).

    CAS  PubMed  Google Scholar 

  95. Di, Q. et al. Impaired cross-activation of β3 integrin and VEGFR-2 on endothelial progenitor cells with aging decreases angiogenesis in response to hypoxia. Int. J. Cardiol. 168, 2167–2176 (2013).

    PubMed  Google Scholar 

  96. Yougbaré, I. et al. Maternal anti-platelet β3 integrins impair angiogenesis and cause intracranial hemorrhage. J. Clin. Investig. 125, 1545–1556 (2015).

    PubMed Central  PubMed  Google Scholar 

  97. Yang, H. et al. Fibrinogen and von Willebrand factor‐independent platelet aggregation in vitro and in vivo. J. Thrombosis Haemost. 4, 2230–2237 (2006).

    CAS  Google Scholar 

  98. Ley, K., Rivera-Nieves, J., Sandborn, W. J. & Shattil, S. Integrin-based therapeutics: biological basis, clinical use and new drugs. Nat. Rev. Drug Discov. 15, 173 (2016).

    CAS  PubMed Central  PubMed  Google Scholar 

  99. O’Connor, P. Natalizumab and the role of α4-integrin antagonism in the treatment of multiple sclerosis. Expert Opin. Biol. Ther. 7, 123–136 (2007).

    PubMed  Google Scholar 

  100. Nagaraj, R. et al. High density display of an anti-angiogenic peptide on micelle surfaces enhances their inhibition of αvβ3 integrin-mediated neovascularization in vitro. Nanomaterials 10, 581 (2020).

    CAS  PubMed Central  Google Scholar 

  101. Majumder, P. Integrin-mediated delivery of drugs and nucleic acids for anti-angiogenic cancer therapy: current landscape and remaining challenges. Bioengineering 5, 76 (2018).

    CAS  PubMed Central  Google Scholar 

  102. Li, Y. et al. An integrin-based nanoparticle that targets activated hepatic stellate cells and alleviates liver fibrosis. J. Controlled Release 303, 77–90 (2019).

    CAS  Google Scholar 

  103. Wang, Y. et al. Intravenous treatment of choroidal neovascularization by photo-targeted nanoparticles. Nat. Commun. 10, 1–9 (2019).

    Google Scholar 

  104. Kossen, K. et al. IDL-2965: a selective, highly-potent, oral Integrin antagonist for IPF. Eur. Respir. J. 54, PA5374 (2019).

  105. Li, J. et al. Novel pure αVβ3 integrin antagonists that do not induce receptor extension, prime the receptor, or enhance angiogenesis at low concentrations. ACS Pharmacol. Transl. Sci. 2, 387–401 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  106. Tan, S.-M. The leucocyte β2 (CD18) integrins: the structure, functional regulation and signalling properties. Biosci. Rep. 32, 241–269 (2012).

    CAS  PubMed  Google Scholar 

  107. Fagerholm, S. C., Guenther, C., Asens, M. L., Savinko, T. & Uotila, L. M. Beta2-integrins and interacting proteins in leukocyte trafficking, immune suppression, and immunodeficiency disease. Front. Immunol. 10, 254 (2019).

  108. Humphries, M. J. Integrin structure. Biochem. Soc. Trans. 28, 311–339 (2000).

    CAS  PubMed  Google Scholar 

  109. Luo, B.-H., Carman, C. V. & Springer, T. A. Structural basis of integrin regulation and signaling. Annu. Rev. Immunol. 25, 619–647 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  110. Xiong, J. P. et al. Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Science 296, 151–155 (2002).

    CAS  PubMed  Google Scholar 

  111. Takagi, J., Petre, B. M., Walz, T. & Springer, T. A. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell 110, 599–511 (2002).

    CAS  PubMed  Google Scholar 

  112. Ferrara, N., Gerber, H.-P. & LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 9, 669–676 (2003).

    CAS  PubMed  Google Scholar 

  113. Abhinand, C. S., Raju, R., Soumya, S. J., Arya, P. S. & Sudhakaran, P. R. VEGF-A/VEGFR2 signaling network in endothelial cells relevant to angiogenesis. J. Cell Commun. Signal. 10, 347–354 (2016).

    PubMed Central  PubMed  Google Scholar 

  114. Yang, W. J. et al. Paxillin regulates vascular endothelial growth factor A-induced in vitro angiogenesis of human umbilical vein endothelial cells. Mol. Med. Rep. 11, 1784–1792 (2015).

    CAS  PubMed  Google Scholar 

  115. Sin, C. C. Effect of PI3K δ Isoform on the Expression of Integrin Beta 3 and p130cas in Glioblastoma Multiforme. MSc thesis, Hong Kong Polytechnic Univ. (2016).

  116. Munger J. et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: A mechanism for regulating pulmonary inflammation and fibrosis. Cell. 96, 319–328 (1999).

    CAS  Google Scholar 

  117. Acharya M. et al. αv Integrin expression by DCs is required for Th27 cell differentiation and development of experimental autoimmune encephalomyelitis in mice. J Clin Invest. 120, 4445–4452 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  118. Liu S. et al. Expression of integrin β1 by fibroblasts is required for tissue repair in vivo. J Cell Sci 123, 3674–3682 (2010).

    CAS  PubMed  Google Scholar 

  119. Reynolds L. et al. Accelerated re-epithelialization in β 3-integrin-deficient-mice is associated with enhanced TGF-β1 signaling. Nat Med. 11, 167–174 (2005).

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

  1. Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA

    Olachi J. Mezu-Ndubuisi

  2. Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA

    Akhil Maheshwari

Authors

  1. Olachi J. Mezu-Ndubuisi

    You can also search for this author in PubMed Google Scholar

  2. Akhil Maheshwari

    You can also search for this author in PubMed Google Scholar

Contributions

Substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; drafting the article or revising it critically for important intellectual content; and final approval of the version to be published: both authors.

Corresponding author

Correspondence to Olachi J. Mezu-Ndubuisi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and Permissions

About this article

Cite this article

Mezu-Ndubuisi, O.J., Maheshwari, A. The role of integrins in inflammation and angiogenesis. Pediatr Res 89, 1619–1626 (2021). https://doi.org/10.1038/s41390-020-01177-9

Download citation

  • Received: 19 June 2020

  • Revised: 18 August 2020

  • Accepted: 10 September 2020

  • Published: 07 October 2020

  • Issue Date: May 2021

  • DOI: https://doi.org/10.1038/s41390-020-01177-9

Further reading

What type of proteins are transmembrane proteins?

There are two basic types of transmembrane proteins: alpha-helical and beta barrels. Alpha-helical proteins are present in the inner membranes of bacterial cells or the plasma membrane of eukaryotic cells, and sometimes in the bacterial outer membrane. This is the major category of transmembrane proteins.

What are transmembrane proteins also called?

Transmembrane protein (TP), also known as intact protein, is a type of membrane protein exists in the whole biofilm, that is, transmembrane proteins span from one side of the membrane to another side.

What are transmembrane proteins responsible for?

Function of membrane and protein synthesis in the cell The columnar transmembrane proteins make up about half of the cell membrane by mass and are responsible for the specialized membrane functions. (1) Some transmembrane proteins are junctions connecting and joining two cells together.

What is a transmembrane receptor protein?

Membrane receptors are usually transmembrane proteins. Transmembrane proteins with part of their mass on both sides of the membrane are poised structurally to transmit information from one side of the membrane to the other. The domain of the receptor exposed to the external medium often has a binding site for a ligand.