Next Article in Journal
Impact of Targeted Deletion of the Circadian Clock Gene Bmal1 in Excitatory Forebrain Neurons on Adult Neurogenesis and Olfactory Function
Next Article in Special Issue
Heterogeneity of Integrin αIIbβ3 Function in Pediatric Immune Thrombocytopenia Revealed by Continuous Flow Cytometry Analysis
Previous Article in Journal
Structural Analysis and Design of Chionodracine-Derived Peptides Using Circular Dichroism and Molecular Dynamics Simulations
Previous Article in Special Issue
The RND1 Small GTPase: Main Functions and Emerging Role in Oncogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

β2 Integrins—Multi-Functional Leukocyte Receptors in Health and Disease

Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(4), 1402; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041402
Submission received: 6 January 2020 / Revised: 11 February 2020 / Accepted: 14 February 2020 / Published: 19 February 2020
(This article belongs to the Special Issue Integrin Signaling and Human Pathologies 2.0)

Abstract

:
β2 integrins are heterodimeric surface receptors composed of a variable α (CD11a-CD11d) and a constant β (CD18) subunit and are specifically expressed by leukocytes. The α subunit defines the individual functional properties of the corresponding β2 integrin, but all β2 integrins show functional overlap. They mediate adhesion to other cells and to components of the extracellular matrix (ECM), orchestrate uptake of extracellular material like complement-opsonized pathogens, control cytoskeletal organization, and modulate cell signaling. This review aims to delineate the tremendous role of β2 integrins for immune functions as exemplified by the phenotype of LAD-I (leukocyte adhesion deficiency 1) patients that suffer from strong recurrent infections. These immune defects have been largely attributed to impaired migratory and phagocytic properties of polymorphonuclear granulocytes. The molecular base for this inherited disease is a functional impairment of β2 integrins due to mutations within the CD18 gene. LAD-I patients are also predisposed for autoimmune diseases. In agreement, polymorphisms within the CD11b gene have been associated with autoimmunity. Consequently, β2 integrins have received growing interest as targets in the treatment of autoimmune diseases. Moreover, β2 integrin activity on leukocytes has been implicated in tumor development.

1. Introduction

Integrins are evolutionarily conserved heterodimeric transmembrane receptors that consist of an α and a β subunit [1]. Vertebrates possess a total of 24 αβ heterodimeric integrins, and some integrin receptors share the same β subunit. Integrins mediate intercellular as well as cell-matrix interactions [2]. Besides their role as adhesion molecules, some integrins also internalize extracellular material [3]. In general, binding of a ligand to an integrin induces signaling events that may result in cytoskeletal remodeling required, e.g., to confer cell migration [4], and activation/modulation of various signaling pathways [5]. As depicted in Figure 1, some integrins bind laminin, and/or the RGD peptide within their target ligands, whereas others recognize the triple-helical GFOGER sequence in collagens [2]. Some integrins also engage cell surface adhesion receptors (e.g., β2 and β7 integrins), apoptotic cells, and/or multiple other ligands. According to this classification, b1 and integrins as well as αV containing integrins constitute the three largest groups. β1α4, β1α9, all β2, and β7αE integrins are rather specifically expressed by leukocytes [6].
This review aims to highlight the role of β2 integrins in innate and adaptive immune cells since this group of receptors is crucially involved in leukocyte differentiation, activation/polarization, and functional activity. Therefore, impaired activity of β2 integrins results in dysregulated immune responses and severe diseases, including autoimmunity and tumor development [7]. Consequently, β2 integrins have been identified as highly interesting targets for therapy [6].

2. Structure and Activation of β2 Integrins

β2 integrins are heterdimeric surface receptors that are composed of one variable α subunit and a non-covalently bound constant β2 (CD18, gene name; Itgb2) subunit [6]. The α subunits are αL (CD11a, Itgal), αM (CD11b, Itgam), αX (CD11c, Itgax), and αD (CD11d, Itgad). β2 integrins dimerize intracellularly and are subsequently integrated in the cell membrane. Hence, single subunits are not detectable on the cell surface [8]. Since CD18 is expressed abundantly, the expression levels of the different α subunits limit the amount of the corresponding β2 integrin on the cell surface [9].

2.1. The α Subunit

The head of any α subunit is composed of a seven-bladed β propeller motif that is connected via a thigh to the calf-1 (c1) and calf-2 (c2) domain (Figure 2). Calcium-binding EF-hand domains found within last three propeller blades promote ligand binding on the other pole of the propeller upon recruitment of a divalent cation [10]. Between the 2nd and 3rd blade of the β propeller a 200 amino acid I domain (also known as A domain) enables the β2 integrin receptor to bind collagen with its αC helix [11]. The αI domain, expanding the cleft between the β propeller and the αI domain of the α subunit, provides a binding surface that allows interaction with larger ligands. Binding of Mg2+ to the metal ion-dependent adhesion site (MIDAS) motif of the αI domain bridges binding of the β2 integrin to collagen or ICAM (intercellular adhesion molecule) over their negative glutamate-rich residues [12]. A C-terminal glutamate residue of the α1 domain constitutes an intrinsic ligand for the MIDAS on the β1 domain of the integrin β subunit, ensuring its conformational change upon ligand binding. The crystal structure of a metastable αXβ2 (CD11c/CD18) revealed that in a high affinity open form, α1α7 helices (C-terminal α7 of I domain) unwind and bind the β subunit between the β propeller and the βI domain [13]. Blockade of the interaction between the α1 and the β1 domains disrupts conformational changes [14]. Ligand binding to β2 integrin receptors may result in conformational alterations resulting in signal transmission into the cell, which has been termed outside-in signaling [3], conveyed by the βA domain of the β subunit. The C terminal portion of αlα7 reshapes into internal ligand and induces polar interaction between the α and β chains [15]. The functional role of the cytoplasmic tail of the α subunit is still unknown.

2.2. The β Subunit

The β subunit is connected to the cytoskeleton and conveys intracellular signaling. It contains eight extracellular domains: four epidermal growth factor (EGF)-like domains (EI-4), a so-called hybrid (H), a plexin-semaphorin-integrin (PSI), a β tail and a βI domain. The βI domain contains a MIDAS as well, which binds Mg2+ and thus bridges binding to aspartate residues within a ligand or to the glutamate residues within the αI domain. It acts as an allosteric regulator of αI-ligand binding [16]. Moreover, the intrinsic binding of Glu310 (αL) to the MIDAS of the activated βI domain leads to transmission of an activated state from the α to the β subunit, and is required for overall αM β2 activation [17]. Adjacent to the MIDAS, an ADMIDAS motif is located [18]. It constitutes a negative regulator inhibiting activation of the β2 integrin receptor at high Ca2+ concentrations and stabilizes its closed conformation. However, the ADMIDAS may also play a role in receptor activation as Mn2+ competes with Ca2+ for the binding to the ADMIDAS which results in receptor activation as demonstrated for β3 integrins [19]. The β1 domain contains a ligand-associated metal binding site (LIMBS) recruiting Ca2+ [20]. In case of β3 integrins, the LIMBS stabilizes binding of the metal ion at the MIDAS [21] and may play a similar role in case of β2 integrins.

2.3. Activation of β2 Integrins

It is well known that integrins are normally expressed in a low affinity state on the cell surface with a bent closed headpiece (Figure 3). Activation of high-affinity binding and of intracellular signal transduction can occur via extracellular (`outside-in signaling´) or via intracellular signals (`inside-out signaling´) [22]. The inactive state of β2 integrins enables circulation of leukocytes in non-inflamed vessels [23]. As an exception, MAC-1 (macrophage-1 antigen, CD11b/CD18) on macrophages was reported to be in a high affinity state by default [24]. Engagement of an agonist (e.g., chemokines) and the presence of elevated levels of divalent cations result in inside-out signaling, and conformational changes of the extracellular domain of the integrin towards a more open, intermediate to high affinity state with an open headpiece. For the binding of some ligands, like ICAM-1, β2 integrins need to be in the high affinity state.
In comparison to LFA-1 (leukocyte factor 1, CD11a/CD18), inside-out signaling of MAC-1 is less well understood, and is distinctly regulated [25]. It is known for instance that stimulated MAC-1 orchestrates migration of polymorphonuclear granulocytes (PMN) in a MAPK-dependent manner, whereas activated LFA-1 induces phosphoinositide 3-kinase (PI3K)-dependent cell motility [26]. As outlined below, LFA-1-dependent cell migration depends on the small GTPase Rap-1 which mediates inside-out signaling [27].
In response to chemokine binding, Gα/Gβγ-associated PLC (phospholipase C) generates the secondary messengers diacylglycerol (DAG) and IP3 (inositol trisphosphate) (Figure 4). In contrast to DAG, IP3 is soluble and confers release of Ca2+ from the endoplasmatic reticulum. DAG and Ca2+ activate PKC and CalDAG-GEFI (calcium and diacylglycerol-regulated guanine nucleotide exchange factor I) [28]. Further, Rap-1 activation is orchestrated by the regulator of adhesion and polarization enriched in lymphocytes (RAPL) molecule [29] which colocalizes both with active Rap-1 and CD11a. Downstream to RAPL the Ser/Thr kinase Mst1 (Macrophage-Stimulating Protein 1) subsequently binds the cytoplasmic CD11a domain and assures allocation of LFA-1 to the leading edge of the cell as a prerequisite for its migration [30]. RIAM (Rap-1-GTP interacting adaptor molecule) engages both active Rap-1 and CD18 [31] and mediates binding of Talin to CD18 [32]. Talin is necessary for unbending of the two β2 integrin subunits [33]. Kindlin-3 binds CD18 as well [34], and both Talin and Kindlin-3are required for the induction of a high affinity state of the β2 integrin [35]. Talin is necessary and sufficient to induce an intermediate state of β2 integrin affinity and to confer slow rolling of PMN on endothelium [36]. In contrast, in the same study both Talin and Kindlin-3 were reported as essential to confer a high affinity state and migration arrest to to binding of ICAM. More recently, Yago and co-workers reported that chemokine signaling also activated both PIP5Kγ90 (phosphatidylinositol-4-phosphate 5-kinase γ90) and PI3Kγ dependent signaling which cooperated with Rap-1 to achieve an intermediate state of affinity of LFA-1 [37]. In addition, binding of PSGL-1 (P-selectin glycoprotein ligand-1) to selectins as expressed by endothelial cells activated Rap-1 and PIP5Kγ90 signaling via Src kinases as well. Besides Talin and Kindlin-3, also the Rho-GEF Cytohesin-1 was reported to bind CD18 and to be necessary for the high affinity state of LFA-1 [38], which as shown for DC required RhoA activation [39]. The importance of Kindlin-3 for β2 integrin activation and downstream functions is underscored by the observation that in human loss-of-function mutations of Kindlin-3 result in a LAD-I-like phenotype, termed LAD-III [40].
It has been shown that phosphorylation of serine residues near the C-terminus of the α subunit in both LFA-1 and MAC-1 is essential for receptor activation and thereby ligand affinity [41]. Subsequent to LFA-1 activation, the transcriptional activator JAB1 (Jun activating binding protein-1) was described to interact with the cytoplasmic portion of a cross-linked β2 integrin and to accumulate thereafter in the nucleus, where it triggers AP-1(activator protein-1) [42]. AP-1 is also a transcription factor and regulates differentiation, proliferation and apoptosis of the cell [43]. Chemokine-triggered β2 integrin activation can transduce signaling over small GTPases, Rap [27] and/or Rho (Ras homolog gene) [39], Rac-1 (Ras-related C3 botulinum toxin substrate 1) [44], Cdc42 (Cell division control protein 42 homolog) [45], according Rho-GEF such as, CALDAG–GEF [46], DOCK2 (Dedicator of cytokinesis 2) [47], VAV1 (Vav Guanine Nucleotide Exchange Factor 1) [48], kinases, including STK4 (Serine/threonine-protein kinase 4) and SKAP55 (Src kinase-associated phosphoprotein 5) [49], and other signaling proteins like PLD1 (Phospholipase D1) [50] and adaptor proteins as ADAP (Adhesion and degranulation-promoting adaptor protein) [49] and PIP5K1C (Phosphatidylinositol-4-Phosphate 5-Kinase Type 1 Gamma) [51,52]. Altogether, inside-out signaling induces a conformational change of the β2 integrin, leading to a high affinity open state, which results in strong binding of all ligands [53]. Engagement of a ligand conveys outside-in signaling resulting in cytoskeletal rearrangements.

2.4. Ligands of β2 Integrins

As mentioned above, the α subunit of a β2 integrin determines its ligand specificity. In this regard, activated LFA-1 engages ICAM 1-5, JAM (junctional adhesion molecule) 1, and ESM (endothelial cell-specific molecule) 1 [54]. MAC-1 binds numerous cell surface receptors at high affinity state and numerous soluble ligands irrespective of its state of activation. The former group comprises ICAM1-4, VCAM-1 (Vascular cell adhesion protein 1), JAM-3 (Junctional adhesion molecule 3), Thy-1 (Thymus cell antigen 1), RAGE (Receptor for advanced glycation endproducts), DC-SIGN (Dendritic cell-specific ICAM-3-grabbing non-integrin), and CD40L [41]. The list of MAC-1 binding soluble ligands is quite extensive, and includes fibrin(ogen), Factor Xa, platetelet Ib, heparin, polysaccharides, ssDNA, dsRNA, some acute phase proteins, HMGB1 (High mobility group box 1) and denatured proteins among many others as well as apoptotic bodies [55]. Moreover, MAC-1 binds several matrix proteins, like collagen, fibronectin, fibrinogen, vitronectin, Cyr61, and plasminogen. Further, MAC-1 is the primary receptor for complement C3-opsonized pathogens and immune complexes at high affinity, recognizing iC3b, C3dg, and C3d, and mediates their internalization [56,57]. Due to this property, MAC-1 has also been termed complement receptor 3 (CR3). (CR1 is widely expressed by leukocytes, and CR2 is predominantly apparent on B cells [58].) The spectrum of CD11c/CD18 ligands partially overlaps with that of MAC-1, including heparin, polysaccharides, and negatively charged denatured proteins [59]. CD11c/CD18 also binds ICAM1, ICAM4, Thy-1, and VCAM-1. Similar to MAC-1/CR3, CD11c/CD18 also recognizes complement-opsonized material, confers its uptake, and accordingly has been termed CR4 [56]. CD11d/CD18 bears structural similarity with MAC-1 and binds serum (Fibrinogen, Plasminogen) and ECM (Cyr61, Fibronectin, Vitronectin) components as well as cellular surface receptors (VCAM-1) [60] as previously reported for MAC-1. More recently, Yakubenko and co-workers demonstrated that PMN, as the first leukocyte population that infiltrates inflamed tissue, mark a path for subsequently attracted macrophages [61]. Activated PMN released myeloperoxidase (MPO), which in turn induced ROS (reactive oxygen species). ROS oxidized polyunsaturated fatty acids. Thereby, CEP (2-ω-carboxyethyl) are generated which modified proteins of the ECM. The derived CEP adducts were engaged at higher extent by MAC-1 and CD11d/CD18 than non-modified ECM components.

2.5. Expression Pattern of β2 Integrins

β2 integrins are expressed only by leukocytes. LFA-1 is rather ubiquitously expressed [62], whereas MAC-1 is predominantly apparent on myeloid cells, including PMN, monocytes/macrophages, and conventional dendritic cells (DC) [63]. In addition, it has been reported that MAC-1 is also present on the surface of NK (natural killer) cells and fractions of mast cells, B cells, CD8+ T cells, and CD4+ γδ T cells [64]. In mouse, CD11c/CD18 is rather specifically expressed by all DC populations, and accordingly serves as a well-accepted pan-DC marker [65]. Especially in human, CD11c/CD18 is also present on NK cells and lymphocyte subpopulations [66]. In mouse, CD11d/CD18 is expressed by a small fraction of leukocytes, being most abundant on myeloid cell lineages like macrophages and like MAC-1 is upregulated upon inflammation. In human, CD11d/CD18 is highly expressed on NK cells, B cells, and γδT cells as well [67].

3. Cellular Functions of β2 Integrins

As outlined in the following, β2 integrins regulate the differentiation (3.1) and the functional properties of immune cells by exerting and modulating a large array of functions (Figure 5). In case of inflammation, β2 integrins confer cell migration, binding both components of the ECM as well as endothelial cells as a prerequisite tfiltrate inflamed tissue (3.2). β2 integrins play a dominant role for the uptake of opsonized pathogens and immune complexes (3.3). Further, β2 integrins regulate in part the state of cell activation in response to ligand engagement. In addition, β2 integrins also modulate the stimulatory activity of (other) danger signals comprising pathogen-derived molecular patterns like LPS [68] as well as endogenous mediators released in response to inflammation as for example TNF-α [69] (3.4). Moreover, β2 integrins regulate the extent and character of immune responses since they constitute essential components of the immunological synapse between antigen presenting cells (APC) and T cells (3.5.1) as well as between effector immune cells and infected/malignant target cells (3.5.2).

3.1. Cell Differentiation

LFA-1 contributes to the development of common lymphoid progenitors, resulting in lower numbers of thymocytes in CD11a−/− mice [70]. However, in the periphery absolute numbers of T cell and B cells were affected only moderately, which suggested compensatory mechanisms. LFA-1 also regulates the polarization of CD4+ T cells. Both CD18hypo [71] and CD11a−/− [72] mice presented with diminished frequencies of CD4+CD25+ regulatory T cells (Treg), and these conferred diminished suppressive activity on stimulated naïve T cells. Moreover, when stimulated under Treg-promoting conditions, CD11a−/− CD4+ T cells differentiated predominantly to pro-inflammatory Th17 cells. Preferred Th17 differentiation was also observed upon antibody-mediated blockade of CD18 in cocultures of DC with WT Treg [71]. Moreover, LFA-1 was reported as required for expression of the transcriptional silencer BCL6 (B cell lymphoma 6) [73] which in turn is a critical regulator of T follicular helper (Tfh) cells [74]. CD11a−/− mice developed much less Tfh in response to infection than WT mice. In accordance with the role of Tfh to exert B cell help as necessary to induce humoral immune responses and to promote antibody affinity maturation [75], helminth-infected CD11a−/− mice were characterized by diminished Tfh induction, and displayed an attenuated humoral immune response [73].
MAC-1 has been attributed an important role in the differentiation of bone remodeling cells. Bone homeostasis is mediated by the concerted activities of osteoblasts that generate, and osteoclasts that dissolve and absorb bone material [76]. Osteoclast progenitors express MAC-1, and macrophages can transdifferentiate to osteoclasts [77]. CD11b−/− mice presented with bone loss that was associated with higher numbers of osteoclasts as compared with WT (wild type) animals [78]. MAC-1 expressing WT osteoclast progenitors were characterized by lower induction of the osteoclastogenesis transcription factor NFATc1 (nuclear factor of activated T cells, cxytoplasmic 1) in response to RANKL (receptor activator of nuclear factor kappa-Β ligand) than CD11b−/− cells. MAC-1 mediated this effect by down-regulating the expression of RANK, and translocation of BCL6 to the NFATc1 gene promoter. Therefore, MAC-1 may constitute a negative feedback regulator of osteoclastogenesis. CD11d−/− mice were characterized by reduced CD3 and CD28 expression by T cells, and an altered ratio of CD4+ and CD8+ T cells [79]. These alterations may be caused by a lack of CD11d/CD18 expression in the thymus resulting in inaccurate T cell development. The overall importance of β2 integrins for the expansion of HPSC (hematopoietic stem and progenitor cells) was demonstrated in a study by Meng and co-workers [80]. In a coculture system of HSPCs and Kupffer cells, that constitute the liver-resident macrophage population [81], Kuppfer cells promoted HSPC expansion and differentiation to lymphocytes. This was inhibited by antibody-mediated blockade of ICAM-1. Further, NK cells in CD18−/− mice were characterized by partially inhibited differentiation as evidenced by an accumulation of c-kit+ progenitor cells, and NK hyporesponsiveness towards stimulation in vitro [82].

3.2. Migration

β2 integrins are essential for the recruitment of immune cells to sites of inflammation or tissue damage. All four β2 integrins bind proteins of the ECM, and surface receptors involved in intercellular interactions [83]. Therefore, β2 integrins enable a cell to adhere to the endothelium, and to extravasate blood vessels at inflamed sites [84]. As mentioned above, of all β2 integrins T cells express only LFA-1 [62], whereas myeloid cells (PMN, monocytes/macrophages and conventional DC) coexpress both LFA-1 and MAC-1 [63,64].
The relative importance of either of these two β2 integrins for migration of myeloid cells, especially of PMN, has been assessed in a number of studies. Heit and co-workers reported that MAC-1 and LFA-1 conferred chemotaxis of PMN in a ligand-specific manner [85]. While LFA-1 was required for IL-8-directed migration, MAC-1 was necessary for migration towards fLMP (N-Formyl-Met-Leu-Phe). Comparative analysis of CD11a−/−, CD11b−/− and CD18−/− PMN revealed that only WT and MAC-1-, but not LFA-1-deficient PMN engaged ICAM-1 at higher extent in response to stimulation with zymosan [86]. Intravital microscopy of leukocyte attachment to venules of muscles pretreated with TNF-α demonstrated that leukocyte velocities were highest in case of CD18−/− mice in comparison to WT mice and displayed intermediate rates in case of CD11a−/− and CD11b−/− mice [87]. Leukocyte adhesion to TNF-α-stimulated endothelial was at comparably low level in case of CD18−/− and CD11a−/− mice and only somewhat lowered in case of CD11b−/− mice. In a similar experimental approach, MAC-1 activity was identified as necessary and sufficient to confer rolling of PMN [88]. In contrast, rolling of monocytes was regulated by both LFA-1 and MAC-1 under non-inflamed conditions, whereas in response to treatment of muscles with TNF-α, MAC-1 played a more important role. In tissue, monocytes differentiate to macrophages that exert either proinflammatory functions to eradicate pathogens, termed M1 macrophages, or serve to dampen inflammation and to facilitate tissue healing (M2 macrophages) [89]. Recently, Cui and co-workers demonstrated that in vitro polarized M1 macrophages expressed CD11d/CD18 at much higher extent than M2 macrophages [90]. Elevated CD11d/CD18 expression enhanced the adhesion of M1 macrophages to the ECM and attenuated their migratory activity as compared to M2 macrophages both in vitro and in vivo as assessed by adoptive transfer of fluorescence-labeled macrophage populations in mice. Interestingly, in contrast to PMN and monocytes/macrophages, immature DC that engaged ICAM-2 on endothelial cells did not require β2 integrins for transendothelial migration [91]. The Rho-GEF Cytohesin—1 negatively regulates β2 integrin activity by engagement of CD18 [32]. Cytohesin-1 was reported to upregulate RhoA activity in stimulated DC, and both factors accounted for chemokine-induced β2 integrin activation [39]. In agreement, silencing of Cytohesin-1 impaired DC migration. CYTIP (cytohesin-1-interacting protein) sequesters Cytohesin-1 in the cytoplasm and thereby limits its interaction with β2 integrins [92,93]. Several viral pathogens were demonstrated to inhibit CYTIP in infected DC, which resulted in elevated LFA-1 activity, and thereby increased cell adhesion and impaired DC motility [94,95]. Furthermore, DC derived from mice with a knock-in of mutated CD18 containing a defective Kindlin-3 binding site showed a more mature phenotype and migrated at higher extent to draining lymph nodes both under steady state conditions [96] as well as after activation in a model of contact hypersensitivity [97].
In the course of T cell migration, redistribution of activated LFA-1 was reported to require Mst1 that is activated in response to chemokine stimulation and conferred activation of the GEF DENND1C (differentially expressed in normal and neoplastic cells domain 1C) as well as of the actin binding protein VASP1 (Vasodilator-stimulated phosphoprotein 1) [98]. Activated VASP1 mediated prolongation of actin filaments, while DENNDC1 activated the GTPase Rab13. Rab13 and Mst1 engaged LFA-1 and mediated its transport along VASP1-induced actin filaments engaging MyosinVa towards the front end of migrating T cells. Further, Mst1 also activated Myosin IIa which contributed to LFA-1 relocalization in migrating T cells as well [99]. Vesicular transport of LFA-1 from the rear to the cell front was reported to require activity of the small GTPase RhoB, which in turn activated Rab11 [100]. The latter is located in recycling endosomes and controls recycling of endocytosed proteins [101]. Moreover, T cell activation in the course of LFA-1 outside-in signaling activated PKCε that mediated phosphorylation of the Rab GTPase Rab5a [102], which is primarily known as a constituent of endocytic vesicles [103]. Activated Rab5a relocalized to the front of migrating T cells and conferred Rac1 activation [102], known to be necessary for rearrangement of the cytoskeleton, and hence T cell migration [104]. The cystein protease Cathepsin X was demonstrated to negatively regulate the high-affinity state of LFA-1 by cleaving a minor part of the C-terminal end of LFA-1, which resulted in preferential binding of alpha-actinin-1 to LFA-1 [105]. Interaction of the PDZ-binding domain of the proteoglycan Syndecan-2 with LFA-1 was also reported to inhibit the acquisition of a high-affinity conformation and thereby elevated intercellular adhesion [106]. Triggering of plexin D1 by semaphorin 3E inhibited Rap-1, which in turn prevented LFA-1 activation and thereby impaired T cell migration [107]. In human monocytes, chemokine-induced LFA-1 activation was limited by the JAK family member PTPRG (protein tyrosine phosphatase receptor type g) [108].

3.3. Phagocytosis

MAC-1 was the first integrin receptor demonstrated to facilitate phagocytosis [109]. It plays a crucial role in the clearance of pathogens, tumor cells, apoptotic cells and of cellular debris that are opsonized with fragments of complement factor C3 [56]. Although physical interaction of MAC-1 with an FcR (Fc receptor) was never observed in murine immune cells, Jongsta-Bilen and colleagues (2003) demonstrated that in case of murine leukocytes that form a phagocytic cup upon FcR engagement MAC-1 accumulation was observed [110]. Likewise, as mentioned above CD11c/CD18 engages pathogens and other material opsonized with complement C4, and accordingly this β2 integrin receptor was also termed CR4 [57]. Hence, whereas FcR bind antibody-opsonized pathogens, MAC-1/CR3 and CR4 are the most important opsonophagocytic receptors of conventional DC. Moreover, in human PMN FcγRIIIB is constitutively associated with MAC-1 [111]. Similarly, MAC-1 was reported to physically interact with FcγRIIA in human PMN and to amplify calcium-mediated signaling of FcγRIIA/B, which resulted in an enhanced phagocytosis and release of pro-inflammatory cytokines [112]. The authors suggested that binding of immune complexes to the FcR resulted in intracellular rearrangements which in turn conferred release of MAC-1 from the cytoskeletal network and allowed its transfer to the site of phagocytosis [113]. There, MAC-1 could re-anchor to the cytoskeleton, and support FcR-mediated phagocytosis. Thus, several studies have confirmed that MAC-1, aside from its intrinsic phagocytic activity, is required for efficient FcR-mediated phagocytosis [114]. Consequently, MAC-1 may support the inflammatory process by amplifying FcR-mediated signaling, as well as by mediating migration of the leukocytes to the site of inflammation, ROS production, antibody-mediated phagocytosis, and release of pro-inflammatory cytokines.

3.4. β2 Integrin Signaling Events in APC

β2 integrins have been demonstrated to modulate cell signaling in response to TLR (toll-like receptor) 4-mediated stimulation. CD14 is a surface membrane protein predominantly expressed by myeloid cell types which acts as a co-receptor for ligands of TLR2 and TLR4 but is also involved in uptake of inflammatory lipids [115]. CD14 is essential for LPS-triggered endocytosis [116]. CD11b was identified as a constituent of the receptor complex regulating TLR4 entry into bone marrow-derived DC [117]. CD11b deficiency resulted in reduced DC activation via TLR4 triggering by LPS which in turn attenuated the T cell stimulatory capacity of DC. Similarly, Perera and co-workers reported that synergistic activation of MAC-1, CD14, and TLR4 in murine macrophages is necessary for their responsiveness towards LPS [118]. Consequently, CD11b−/− macrophages displayed diminished NF-κB and MAPK signaling in response to LPS. In addition, Mac-1 was identified to serve as a receptor for dsRNA and to facilitate its internalization and thereby yielded enhanced TLR3-dependent signaling in macrophages [119]. Furthermore, internalized dsRNA induced NOX2 (NADPH Oxidase 2) in a TLR3-independent manner, which in turn activated both MAPK and NF-κB.
On the contrary, several studies demonstrated that MAC-1 negatively regulated TLR-triggered inflammatory responses, preventing the onset of inflammation and subsequent tissue damage. In this regard, CD11b−/− bone marrow-derived macrophages were shown to secrete higher levels of IL-6 and TNF-α in response to infection with Mycobaterium bovis Bacillus Calmette–Guerin (BCG) than WT macrophages [120]. DC recognize BCG via TLR2 and TLR4 [121]. Stimulation of TLR9 in CD11b−/− DC yielded increased IL-12p70 production as compared with WT DC [122]. In agreement, Yee and Hamerman reported that CD18−/− murine bone-marrow-derived macrophages and DC were hypersensitive towards various TLR ligands, as reflected by elevated production of IL-12 and IL-6 [123]. Bai and co-workers showed that MAC-1 inhibited TLR9- but not TLR4-induced expression of IL-12 in DC [122]. In case of TLR9 triggering, MAC-1 was required to induce miRNA-146a, which in turn inhibited expression of its genuine target NOTCH1 (neurogenic locus notch homolog protein 1), a well-known IL-12 inducer [124]. DC with a knock-in of CD18 mutated in the Kindlin-3 binding site and thereby inhibited β2 intgerin activity were demonstrated to contain higher levels of activated SYK (spleen tyrosine kinase), which via p38 MAPK activation conferred a more mature DC immuophenotype [96]. In agreement with the proposed anti-inflammatory activity of MAC-1 (under homeostatic conditions), engulfment of apoptotic bodies by human monocyte-derived DC via MAC-1 was reported to diminish their T cell stimulatory activity [125,126]. Moreover, MAC-1 was demonstrated to impair B cell receptor signaling in order to maintain autoreactive B cell tolerance [127] and dampened TLR3-dependent stimulation of NK cells [128].
Engagement of cytokines directly triggers the JAK (Janus kinase)/STAT (signal transducers and activators of transcription) signaling pathway [65]. As analyzed in a myeloid cell line, STAT3 activation in turn activated MAC-1, which resulted in homotypic cell aggregates [129]. Moreover, MAC-1 on human macrophages that bound ICAM-1 inhibited TLR signaling in an indirect manner by promoting expression of IL-10, SOCS (suppressor of cytokine signaling) 3, ABIN-3 (A20-binding inhibitor of NF-κB activation 3), and A20 [97]. It has also been shown that pharmacological activation of MAC-1 on NK cells with leukadherin-1 reduced phosphorylation of STAT-5 in response to IL-12 stimulation and attenuated secretion of IFN- (interferon-) γ, TNF- (tumor necrosis factor-) α, and MIP1- (macrophage inflammatory protein 1-) β [130].

3.5. β2 Integrins in the Interaction of Immune Cells

3.5.1. Interaction of APC and T Cells

Composition of the Immunological Synapse

The term immunological synapse (IS) designates the contact region between an APC and a T cell [131]. On cellular level, the IS structure has been studied most extensively for DC and CD4+ T cells [132]. The central supramolecular activation cluster (cSMAC) of an IS largely contains interacting receptor pairs of APC and T cells that confer antigen presentation (MHC, major histocompatibility complex) and recognition of the MHC/antigen complex by the TCR (T cell receptor) and TCR-associated coreceptors like CD3, CD4, and CD8 [133]. (Detailed graphical overviews of the IS structure, signaling processes initiated by T cell stimulation, and cytoskeletal rearrangements are given in [134] and [135].) In addition, the cSMAC contains receptor pairs required to transmit stimulatory signals from the APC (e.g., CD80, CD86) to antigen-specific T cells (CD28). The peripheral SMAC (pSMAC) among other receptor-ligand pairs is also characterized by a high density of ICAM-1 on the APC and its binding partner LFA-1 on the T cell side [136].

Role of LFA-1 in T Cell Activation

We showed that CD4+ T cells stimulated by DC proliferated at lower extent when lacking LFA-1 and were less prone to polarize towards Th1 [137]. This defect was rescued by additional antibody-mediated TCR stimulation, but not the costimulatory receptor CD28. These observations indicated that LFA-1/ICAM-1 interaction lowered the threshold required for optimal T cell stimulation. In agreement with the elevated threshold of TCR activation in LFA-1-deficient T cells, a normal dose of collagen sufficient to induce CIA (collagen-induced arthritis) in WT mice was insufficient in LFA-1−/− animals [138]. However, a higher dose of antigen compensated for LFA-1 deficiency, inducing CIA. MHCII/antigen complex triggered TCR and ICAM-binding LFA-1 collaborated to trigger ERK-1/2 (Extracellular signal-regulated kinase-½) activation in T cells. Subsequent work showed that low affinity antigens depended at higher extent on LFA-1 than high affinity antigens to overcome the threshold of TCR activation [139]. As outlined above, LFA-1 activity is also required for Treg induction [71,72] and is a negative regulator of Th17 [71] and a positive regulator of Tfh [73] induction.

Regulation of LFA-1 activity on T Cells

Initial binding of MHC/antigen complexes to the TCR was shown to activate LFA-1 affinity by inside-out signaling [140]. Furthermore, an increasing number of adaptor molecules involved in the regulation of LFA-1 affinity and its spatial localization have been identified. Initial triggering of the TCR via the phosphatase SHP- (Src homology region 2 domain-containing phosphatase) 1 activated the adaptor protein CrkII and mediated its redistribution towards the pSMAC [141]. There, active CrkII recruited the GEF C3G, which activated the small GTPase Rap-1 (Ras-associated protein 1). Kondo and co-workers delineated that Rap-1 via Mst1/Mst2 activated NDR (Nuclear Dbf2-Related Kinase) 1 kinase [142]. In APC-binding T cells, Rap-1 activation was mediated in part by PI3Kδ [143]. Binding of semaphorin 3E to its receptor plexin D1 caused inhibition of the small GTPase Rap-1 [107]. This resulted in impaired activation of LFA-1, and attenuated IS formation. Activated NDR1 engaged Kindlin-3 and mediated its translocation towards the cSMAC [142]. The LFA-1 binding coactivator Kindlin-3 is defective in LAD-III patients [40]. T cells of a LAD-III patient in contrast to WT T cells showed no T cell spreading on surface-immobilized ICAM-1 when coincubated with DC [144]. Therefore, the authors concluded that Kindlin-3 transmitted TCR-triggered LFA-1 activation.

Cytoskeletal Rearrangements in the Course of T Cell Stimulation

A growing number of studies has highlighted the role of dynamic reorganization of the actin cytoskeleton in the context of IS formation and stabilization with the TCR and LFA-1 as important regulatory nodes [145]. Engagement of the TCR by MHC/antigen complexes induced F-actin relocalization from the cSMAC towards the pSMAC, yielding conformation-dependent activation of LFA-1. Binding of activated LFA-1 to ICAM-1 on the APC was observed to reduce the centripetal flow of F-actin from the cSMAC. Initial engagement of TCR was shown to result in TCR clustering which in turn caused recruitment of WASp (Wiskott-Aldrich syndrome protein) and Arp2/3 (actin-related protein-2/3) yielding actin polymerization [146]. Formins support actin polymerization by elongating actin filaments and inducing actin arcs within the IS [147]. Inhibition of Formins impaired TCR clustering within the cSMAC. TCR-associated Lck (Lymphocyte cell-specific protein tyrosine kinase) activates Crk-associated substrate lymphocyte-type (Cas-L), a force-sensing protein, which in turn mediated arrangement of TCR micro-clusters within the cSMAC [148]. In addition, activated CasL contributed to LFA-1 activation within the pSMAC. Caveolin-1 was required for spatial redistribution of LFA-1 towards the pSMAC [149]. Besides, the tyrosine phosphatase PRL-1 (phosphatase of regenerating liver 1) was demonstrated to be delivered aside with CD3ζ-containing vesicles towards the IS, and to colocalize with the TCR, especially CD3ε, and LFA-1 [150]. Pharmacological inhibition of PRL-1 affected actin rearrangements and IL-2 production.
LFA-1 is anchored to the underlying actin cytoskeleton by various proteins, including the actin-binding protein l-plastin in case of T cells [151]. Inhibition of L-plastin activity impaired LFA-1 redistribution, affected T cell/APC interaction, and resulted in attenuated T cell proliferation. Transgelin 2 (TGLN2) is predominantly expressed in T cells and served to stabilize cortical F-actin and at the same time engaged LFA-1 [152]. TGLN2 counteracted F-actin severing Cofilin activity. Consequently, TGLN2-deficient T cells formed unstable IS. LFA-1 colocalized with Formin-containing actin arcs, and Formin inhibition interfered with LFA-1 arrangement within the pSMAC, which caused attenuated IS stability and TCR signaling [147]. The cytoskeletal protein SPTAN1 (spectrin alpha, non-erythrocytic 1) also colocalized with LFA-1 in the course of IS formation [153]. siRNA-mediated SPTAN1 deficiency resulted in impaired adhesion of T cells to APC, and disturbed IS formation. Furthermore, the focal adhesion proteins paxilin, Talin, and vinculin colocalized with ICAM-1 engaging LFA-1 [154]. Of note, Talin and vinculin were required for inhibition of the centripetal F-actin flow as induced by TCR engagement, associated with attenuated tyrosine phosphorylation. Thereby, LFA-1, on one hand, is necessary for T cell/APC adhesion but, on the other hand, also dynamically regulates the extent/duration of TCR activation by modulating actin rearrangements within the IS [155].

β2 integrin Activity and Cytoskeletal Rearrangements on the APC Side

T cell priming also requires dynamic rearrangements of the actin cytoskeleton on the side of the interacting APC. WASp-deficient DC displayed diminished stability of the Arp2/3 actin filament network within the forming IS which resulted in lower accumulation of MHCII and ICAM-1 [156]. Rho signaling controls the activity of Cofilin that severs F-actin to enable dynamic cytoskeletal rearrangements [157]. We demonstrated that DC which lacked the Rho inhibitor Myosin IXB displayed strongly diminished Cofilin activity and contained less F-actin within the IS [158]. This was associated with altered DC/T cell contact duration and attenuated T cell proliferation within a 3D collagen micro-environment. The actin binding proteins moesin and α-actinin-1 bind the intracellular part of ICAM-1 and mediate its clustering within the IS to enable engagement of LFA-1 on the T cell side [159]. DC with mutated ICAM-1 lacking the cytoplasmic region initiated less IS and were poor T cell activators. Concerning the role of β2 integrins expressed by APC in the context of T cell activation, we reported for conventional DC that MAC-1 when activated by divalent cations attenuated CD4+ T cell proliferation [24]. In that study, we also demonstrated that inhibition of MAC-1 on macrophages using a blocking antibody enhanced their T cell stimulatory capacity. These findings confirmed the overall inhibitory activity of MAC-1 on APC. Furthermore, we showed that forced activation of LFA-1 on conventional DC by siRNA-mediated inhibition of LFA-1 binding CYTIP also increased CD4+ T cell stimulation [93]. DC with a non-functional Kindlin-3 binding site within the cytoplasmatic part of CD18 were characterized by elevated expression of surface activation markers and of IL-12 both at unstimulated state and after stimulation and consequently exerted stronger Th1 induction [96]. Altogether, these findings suggested that LFA-1 and MAC-1 on the DC surface may modulate the extent of adaptive immune responses. It is still unknown which counter-receptors on T cells are contacted by LFA-1/MAC-1 on the DC.

3.5.2. Leukocyte/Target Cell Interaction

An IS also forms at the interface between two types of immune cells or between an (activated) immune cell and a target cell. Activated CD8+ cytotoxic T lymphocytes (CTL) were not able to stably adhere to an antigen-presenting tumor cells when CTL were pre-incubated with galectins [160] that bind cell surface glycans and are generated at high extent by tumor cells [161]. Impaired engagement of tumor cells by galectin-covered CTL was associated with impaired redistribution of LFA-1 to the CTL/tumor cell contact site. Antibody-mediated blockade of LFA-1 prevented the formation of such an IS as well. A decisive role of LFA-1 activity for CTL/tumor cell IS formation was confirmed by Wabnitz and co-workers who observed that oxidation-mediated hyperactivation of L-plastin which binds LFA-1 arrested the CTL/tumor cell contact [151]. This in turn attenuated overall CTL killing activity. Forced over-expression of the dual actin and LFA-1 binding protein TGLN2 in CTL yielded stronger killing activity in case of ICAM-1-expressing but not ICAM-deficient antigen-presenting tumor cells [162]. This observation suggested TGLN2-dependent inside-out activation of LFA-1. As mentioned above, TGLN2 was reported as well to stabilize F-actin at the pSMAC enabling LFA-1 mediated IS stabilization [152].

4. Pathophysiological Role of β2 Integrins in Human

Leukocyte adhesion deficiency (LAD) syndromes comprise a group of rare autosomal recessive disorders with an incidence of <1:10,000,000 (nearly 300 reported cases) [163]. In case of LAD-I, the molecular cause is impaired β2 integrin activity due to mutations within the CD18 gene [7,164]. LAD-I patients suffer from recurrent infections of bacterial and fungal origin, resulting in periodontitis, tooth loss, impaired wound-healing as well as severe leukocytosis, splenomegaly, and autoimmune symptoms [165,166]. In the severe form most LAD-I patients die before their 5th year of life, in a moderate form (5% to 15% residual CD18 activity) patients have a high chance of mortality between the 2nd and 4th life decade due to chronic infections [167,168]. CD18 deficiency described in dog and cattle presents with PMN dysfunction and recurrent infections of bacterial origin and thus resembles LAD symptoms reported for humans, which indicates that β2 integrin-associated pathology is conserved among mammalian species [169]. So far, two major blood cell types were considered to contribute largely to this phenotype, namely PMN and platelets, which are critical for the management of bacterial infections and rapid wound-healing, respectively.
The LAD-II syndrome, also known as congenital disorder of glycosylation type IIc, is characterized by symptoms similar to those noted in LAD-I, i.e., recurrent bacterial infections including pneumonia, periodontitis, and otitis media accompanied by leucocytosis [170]. The cause of LAD-II is a deficiency of the GDP-fucose transporter and consequently a defect in the synthesis of Sialyl-Lewis X, a P- and E-Selectin binding carbohydrate that is important for leukocyte tethering and rolling along the endothelium [60,171]. The LAD-III syndrome, also known as LAD-I variant’, is caused by mutations in FERMT3 (Fermitin Family Member 3)/Kindlin-3 that are important for the inside-out signaling and activation of β2 integrins. Thus, the adhesive function of leukocytes and platelets in LAD-III patients is impaired and cells cannot migrate efficiently [172,173,174]. So far, most research has focused on the role of PMN in LAD-associated pathologies. However, by now, other leukocyte populations were shown to play a crucial role in these maladies as well, especially in case of LAD-I and LAD-III.

5. Mouse Models to Study Functions of Distinct β2 Integrins

Even though LAD syndromes are quite rare, investigation of their pathomechanisms provides insight into fundamental immune processes and may serve to develop new immunomodulatory therapies. By now, several mouse models intended to reflect the LAD-I phenotype have been developed. CD18 knock-out mice have been generated first in the laboratory of A. L. Beaudet [175]. For this, an insertion mutation was introduced into the CD18 gene locus employing a homologous recombination approach in embryonic stem (ES) cells. Due to cryptic promoter activity exerted by targeting vector elements, low residual CD18 gene expression occurred which resulted in a hypomorphic CD18 allele (CD18hypo). The homozygous offspring displayed 2% to 16% of WT CD18 expression and was viable and fertile. CD18hypo mice showed mild granulocytosis, an impaired inflammatory response to chemical peritonitis, and a delay in the rejection of cardiac transplants as well as erythrosquamous skin plaques that are strikingly reminiscent to psoriasis [176]. Thus, the CD18hypo mouse model is well-suited to study psoriasis [177,178].
Subsequently, a full CD18 knockout mouse was generated [179]. The phenotype of CD18−/− mice was much more severe as compared to CD18hypo mice. About one third of CD18−/− offspring died perinatally, and those that survived infancy developed extended facial and submandibular ulcerative dermatitis. Inflamed lesions contained lymphocytes but very few PMN, suggesting that migration of these cells was impaired. CD18−/− mice developed granulocytosis, splenomegaly, and lymphadenopathy. They had about 10-fold increased serum IgG levels, and elevated IL-3 and IL-6 serum levels as compared with WT mice. The CD18−/− mouse model resembles therefore a severe form of the LAD-I syndrome in humans and has been explored in the context of various pathologies like psoriasis [180], wound healing [181], diabetes [182], carditis [183,184], osteoporosis [185], and infections [186].
In order to assess the specific role of the different β2 integrins for immune functions, transgenic mice with constitutive deletion of either α subunit were generated. CD11a−/− mice displayed mild neutrophilia and cleared bacterial infections inefficiently [187,188]. LFA-1-deficient mice failed as well to reject tumor xenografts and to respond to alloantigens due to the requirement of LFA-1 for IS formation and subsequent TCR signaling amplification [54]. In addition, LFA-1 was necessary for adhesion of T cells to infected and malignant cells and thereby for their killing [62]. CD11b−/− mice were characterized by diminished PMN activation during inflammation and defective T cell proliferation in response to bacterial infection as a consequence of MAC-1 deficiency [189,190,191]. Interestingly, CD11b−/− PMN were less prone to apoptosis [192]. Further, CD11b−/− mice were more susceptible to develop autoimmune diseases [193]. In addition, CD11b−/− mice were protected from thrombosis in response to injury as MAC-1 engagement of platelet GPIbα results in thrombus formation [194]. In some infection models CD11c−/− mice showed an aggravated course of disease due to the requirement of CD11c/CD18 (CR4) for uptake of C4-opsonized pathogens for the induction of sustained adaptive immune responses [56,109,195,196]. CD11d-deficient mice in some cases displayed an aggravated course of infectious diseases [79].
Taken together, all four β2 integrins exert distinct functions, but display a functional overlap, such as migration and adhesion to inflamed endothelium. Consequently, the different β2 integrins can compensate for the lack of each other to some extent. None of the β2 integrin α chain (CD11a-CD11d) deficient mouse strains resembled the disease phenotype of CD18hypo [175] and CD18−/− [62] mice. In the following section, the pathophysiological role of β2 integrins for control of infections, the induction and course of autoimmune diseases and tumor progression, as well as therapeutic β2 integrin targeting approaches are discussed in more detail.

5.1. Functions of β2 Integrins in Infections

So far, the inability of the immune system of LAD-I patients to control infectious diseases has been largely attributed to functional defects of PMN [197] that constitute the first line of cellular innate host defense against pathogens [198] and of monocytes/macrophages [199]. As outlined above, β2 integrins essentially contribute to the immune functions of myeloid cells, comprising (i) chemokine-induced transendothelial migration due to engagement of endothelial ICAM by LFA-1 and MAC-1 [83]; (ii) recognition of complement-opsonized pathogens via MAC-1/CR3, and CR4 (CD11c/CD18) resulting in both cell activation and pathogen clearance via phagocytic uptake [56,200]; and (iii) cell activation by infection-induced cytokines or (pathogen-derived) ligands that engage MAC-1 and CD11d/CD18, which in turn initiates production of extracellular mediators that kill pathogens [200]. Besides, for canine PMN, LFA-1-mediated binding to ICAM-1 was shown to be sufficient to trigger release of hydrogen peroxide [201], which constitutes one of the various pathogen-killing mechanisms of myeloid cells [200]. In contrast to LFA-1-induced respiratory burst, MAC-1 mediated release of hydrogen peroxide, on one hand, required stimulation by chemokines but, on the other hand, induced much higher levels of hydrogen peroxide [201]. The role of β2 integrins in infections is outlined in the following.

5.1.1. Viral Infections

In a HSV-1 (herpes simplex virus type 1) ocular infection model, CD11c−/− mice showed lower virus titers than WT animals [202]. Attenuated viral loads were associated with stronger expression of IFNI (interferon type 1) and a higher frequency of virus antigen-specific CD8+ T cells in CD11c−/− animals. So far, CR4 (CD11c/CD18) has not been associated with recognition/uptake of HSV-1 by DC, which is in agreement with the finding that HSV-1 has evolved strategies to counteract opsonization by complement [203]. It remains to be shown whether CD11c deficiency partially prevented infection of DC by HSV-1, and thereby HSV-1-induced inhibition of DC activation.

5.1.2. Bacterial Infections

In a sepsis model CD11a−/−, mice were partially protected from lethal shock in response to administration of low doses of LPS [204]. The authors attributed the increased survival of CD11a−/− mice to LPS-induced production of anti-inflammatory IL-10 by macrophages. This observation suggests that LFA-1 limits IL-10 production in myeloid cells. The role of LFA-1/ICAM-1 interaction in the context of infection-induced PMN infiltration was assessed in a model of aerosolized LPS application [205]. Here, PMN recruitment to the lung was roughly halved both in case of CD11a−/− and ICAM-1−/− mice. Similar effects were noted in WT mice when using blocking antibodies for either surface receptor. In a model of polymicrobial sepsis, lung infiltration of PMN was comparable in CD11b−/− and WT mice [206]. However, CD11b−/− mice were characterized by higher bacterial counts and stronger systemic inflammation, indicative of attenuated killing activity of MAC-1 deficient leukocytes. Mast cells have been reported to confer resistance towards acute septic peritonitis [207]. They were shown to express MAC-1 as well, and CD11b−/− mice contained less mast cells as assessed in various organs [208]. Hence, the elevated mortality of CD11b−/− mice in this disease model has been attributed in part to the reduced number and diminished functional activity of mast cells [207].
LAD-I patients with pneumonia displayed aggravated pulmonary PMN infiltration [209]. Similarly, Mizgerd and co-workers reported that infection of mice with Streptococcus pneumonia yielded strong PMN infiltration also in CD18−/− mice [210]. In contrast, intratracheal administration of Escherichia coli and Pseudomonas aeruginosa resulted in strongly diminished pulmonary infiltration of PMN in CD18−/− mice as compared with infected WT animals [211]. These findings suggest that β2 integrin deficiency may be compensated by other adhesion receptors in a disease-specific manner. Similar to CD18−/− mice, CD11b−/− mice infected with S. pneumoniae contained elevated numbers of pulmonary PMN as compared to infected WT mice but at the same time showed higher bacterial burden and stronger inflammation in lung than noted for WT mice. Again, these findings suggested functional impairment of CD11b−/− leukocytes to kill pathogens. However, infection of CD11b−/− mice with Mycobacterium tuberculosis yielded comparable bacterial burden, cytokine production, and development of granulomatous lesions as compared with WT animals [212].
As an immune evasive strategy, several pathogens like Porphyromonas gingivalis, the main inducer of periodontitis [213], bind CR3 [214] to prevent IL-12 induced pathogen-specific Th1 responses [215]. In agreement, CD11b−/− mice infected with P. gingivalis contained higher serum levels of IL-12 and Th1-associated IFN-γ than infected WT mice. In accordance, CD11b−/− mice cleared the pathogen more efficiently than WT animals [216]. In contrast, CD11a−/− mice were characterized by periodontal bone loss [165] as also observed for LAD-I patients [166] as a consequence of infection with Porphyromonas gingivalis. Besides impaired infiltration of PMN required to control the infection, high levels of IL-17 were considered responsible for the aggravated course of periodontitis [165]. Antibody-mediated neutralization of IL-17 or the APC-derived Th17 promoting cytokine IL-23 attenuated the course of disease in CD11a−/− mice. It is conceivable that the intrinsic property of LFA-1-deficient CD4+ T cells to differentiate towards Th17 may explain this phenotype in part.
Listeria monocytogenes is an intracellular pathogen which causes listeriosis in immunocompromised patients [217]. CD18−/− [218] and CD11a−/− [219] mice presented with higher survival after systemic administration of L. monocytogenes than observed for WT mice. This was associated with lower bacterial burden in liver and spleen and, accordingly, attenuated formation of necrotic lesions. In both knockout strains infection with L. monocytogenes caused elevated serum levels of G-CSF (granulocyte-colony stimulating factor) known to promote PMN differentiation [220]. L. monocytogenes-infected CD18−/− mice also displayed enhanced serum contents of the innate proinflammatory cytokine IL-1β [218]. In likewise infected CD11a−/− mice, numbers of liver-infiltrating PMN were elevated, and IL-17 serum levels were enhanced [219]. At early time points after infection with L. monocytogenes, liver PMN of CD11a−/− mice generated highly increased levels of IL-12, which in turn induced IFN-γ production by NK cells [221]. In WT mice, antibody-mediated blockade of CD11b prior to infection with L. monocytogenes caused enhanced liver infiltration of PMN and clearance of the pathogen by PMN and Kupffer cells [222]. The extent of CD8+ T cells responses in mice deficient for α subunits of β2 integrins (CD11a-CD11c) in response to infection with L. monocytogenes was assessed in a comparative study [186]. Only in CD11a−/− mice significant differences as compared with WT animals were observed. In these mice the primary CTL response was strongly attenuated, whereas the generation of central memory CD8+ T cells remained largely unaltered.
CD11c−/− mice presented with an aggravated form of Lyme carditis upon infection with the spirochete Borrelia burgdorferi, which was most likely caused by an increased infiltration of macrophages [223]. Thus, CR4 (CD11c/CD18) may be important for the clearance of infections mediated by pathogens that are opsonized by C4 [56,109,195,196]. In a peritoneal Salmonella Typhimurium infection model, CD11d−/− mice were characterized by diminished peritoneal infiltration of leukocytes and accordingly elevated bacterial loads [224]. Ex vivo analysis confirmed attenuated phagocytic activity of CD11d−/− macrophages, accompanied by increased cell death in the course of the inflammatory response, termed pyroptosis [225].

5.1.3. Fungal Infections

Immunocompromised human, including LAD-I patients, often suffer from pulmonary infections with Aspergillus fumigatus [226]. Recently, we showed that lungs of CD11b−/− mice infected with A. fumigatus contained higher numbers of PMN than observed for WT animals [190]. However, lungs of infected CD11b−/− mice presented with a higher fungal burden and contained less innate proinflammatory mediators than apparent in infected WT mice. In addition, CD11b−/− PMN exerted lower phagocytic activity on complement-opsonized A. fumigatus conidiae. The latter finding is in agreement with a report of Gazendam and co-workers demonstrating that in human CR3-dependent phagocytotic uptake of complement-opsonized A. fumigatus conidiae is the primary mechanism for fungal killing [226]. However, A. fumigatus-triggered PMN activation also yielded release of NET (neutrophil extracellular trap) [227] that are largely composed of decondensed chromatin, and trap pathogens [228]. Clark and co-workers revealed that A. fumigatus cell extracts which stimulated PMN via CR3 also induced ROS production and ROS-dependent NETosis [229]. Like invasive pulmonary aspergillosis, invasive candidiasis frequently occurs in immunocompromised humans [230]. Candida albicans is cleared by CR3-mediated phagocytosis [200], NETosis [231] as well as ROS [232]. Hence, in a mouse model of candidiasis CD11b−/− mice displayed attenuated PMN killing activity and increased fungal burdens [233].

5.1.4. Metazoan Parasites

Leishmania are obligate intracellular unicellular parasites that enter macrophages and cause severe skin lesions [234]. In experimental leishmaniasis, footpads of CD18−/− mice infected with Leishmania major were not infiltrated by PMN, in contrast to infected WT animals [235]. In vitro analysis revealed that CD18−/− macrophages phagocytosed complement-opsonized L. major at lower extent than WT macrophages, which may be explained by lack of MAC-1/CR3. In that study, scavenger receptors that engage a broad range of extracellular compounds including pathogens [236] were shown to confer L. major uptake. Further, CD18−/− macrophages were unable to generate NO [235] which contributes to L. major killing [237]. T cells derived from L. major-infected CD18−/− mice poorly proliferated upon restimulation, which is in agreement with the pronounced role of LFA-1 for T cell activation [62]. Similar to other pathogens, L. major was shown to limit IL-12 production by APC via binding to MAC-1/CR3 [238]. In line, L. major-infected CD11b−/− mice were characterized by a milder course of cutaneous leishmaniasis [239]. It is tempting to speculate that the lack of CR3 prevented inhibitory effects of Leishmania engagement on IL-12 production which is required to mount a Th1-biased anti-Leishmania immune response [240].
Infection of CD11d−/− mice with the malaria pathogen Plasmodium berghei induced less systemic inflammation and attenuated lethality as compared to infected WT animals [241]. However, the advantage of survival of P. berghei-infected CD11d−/− mice was not associated with differences in parasite load. Further, although Th1-biased immune responses are crucial for control of malaria in the acute phase [242], serum of P. berghei-infected CD11d−/− mice contained less amounts of the Th1-promoting cytokine IL-12 than observed for infected WT animals [241]. Hence, the mechanisms that confer an attenuated course of malaria in CD11d−/− mice are unclear so far. CD11d deficiency also attenuated MA-ARDS (malaria-associated acute respiratory distress syndrome) [243]. Infected CD11d−/− mice displayed less lung infiltration by leukocytes, lung inflammation and airway hyper-responsiveness than infected WT animals.
Altogether, these findings suggest that with regard to migratory activity, loss of either LFA-1 or MAC-1 in myeloid cells may be compensated by the other β2 integrin [87]. In addition, the activity of β2 integrins as such may be dispensable in some types of infections, presumably due to partial compensation by other adhesion receptors [244,245,246]. Anyway, loss of MAC-1/CR3 on one hand may diminish pathogen clearance by phagocytic uptake [56] and also attenuate other killing mechanisms of myeloid cells [201,235]. However, a number of pathogens exploits MAC-1/CR3 induced inhibition of IL-12 production in APC and thereby sustained pathogen-specific Th1-biased immune responses [216]. Therefore, the net outcome of MAC-1/CR3 deficiency concerning the course of an infection may depend on the relative functional importance of this β2 integrin for innate and adaptive immunity. Besides, it is noteworthy that also engagement of LFA-1 by some pathogens results in impaired IL-12 production [221]. Furthermore, pathogens have developed additional immune evasion strategies based on modulation of β2 integrin activity. For example, infection of DC by HSV-1 [94] and cytomegalovirus [95] impaired their migratory activity due to virus-induced downregulation of CYTIP-1. This resulted in enhanced LFA-1 activity, and thereby increased cell adhesion. We have shown that active LFA-1 on DC impaired their T cell stimulatory capacity [93]. It is conceivable that lowered migration (to secondary lymphoid organs) and attenuated T cell activity of infected DC may cause impaired adaptive pathogen-specific immune responses.

5.2. Functions of β2 Integrins in Autoimmunity

A growing body of research suggests that β2 integrins play an important role in tolerance induction [247,248,249] and suppression of inflammation [83,250,251]. Already decades ago, it was noted that LAD-I patients suffer not only from bacterial infections but also from renal or intestinal autoimmune diseases [252,253], and some of them presented type 1 diabetes or autoimmune cytopenia after hematopoietic stem cell transplantation [254]. Likewise, CD18−/− mice are characterized by chronic dermatitis and splenomegaly, which indicates a circulating inflammation [179]. These findings may be explained by a tolerance-promoting role of β2 integrins. As outlined above, LFA-1 is required for the differentiation and suppressive activity of Treg, and its deficiency actually promoted induction of proinflammatory Th17 cells [71,72]. Furthermore, MAC-1 was shown to be required for the induction of peripheral tolerance by suppressing IL-6 secretion and subsequent Th17 differentiation in a model of orally induced tolerance in mouse [255]. Furthermore, MAC-1 maintained autoreactive B cell tolerance by inhibiting B cell receptor signaling [127]. Besides, we have reported that activated LFA-1 on DC [93] and MAC-1 on DC and macrophages [24] limited their T cell stimulatory capacity. Most studies have addressed the role of LFA-1 and MAC-1 for the onset and course of autoimmune diseases in various mouse models. The main observations of according studies are summarized in Table 1.

5.2.1. LFA-1

LFA-1 plays a crucial role in transendothelial migration of activated T cells and stabilizes the APC/T cell contact, thereby strengthening TCR signaling [256]. Inducible EAE (experimental autoimmune encephalomyelitis) is a well-established mouse model that recapitulates many aspects of multiple sclerosis (MS) [257]. For induction of EAE, mice are immunized with a protein of the neuronal myelin sheath, e.g., MOG (Myelin oligodendrocyte glycoprotein), in combination with an adjuvant. Both in MS and EAE the autoinflammatory phenotype is mediated predominantly by CD4+ Th17 (T helper cell type 17) cells. EAE can also be induced by transfer of CD4+ T cells from a mouse that has developed EAE (encephalitogenic T cells) to a naive one. CD11a−/− mice presented with a reduced frequency of immuno-suppressive Treg fraction in the CNS, which resulted in more severe EAE after immunization [258]. Likewise, CD18 expression was found to be necessary for Treg induction [259,260]. Thus, on one hand transfer of encephalogeneic WT T cells to CD11a−/− mice—lacking Treg—caused a fatal course of the EAE [261]. On the other hand, transfer of encephalogeneic T cells from CD11a−/− mice, immunized to develop EAE, to WT mice resulted in an attenuated course of EAE, which confirms that T cells require LFA-1 to get primed and to mediate an inflammatory response [261,262].
Concerning the role of LFA-1 in other autoimmune manifestations in humans, an elevated expression of LFA-1 on T cells was shown to correlate with the severity of systemic sclerosis, systemic lupus erythematosus (SLE) [263], rheumatoid arthritis (RA), and autoimmune thrombocytopenia [264,265]. In order to suppress T cell priming and effector functions, antibody-mediated blockade of CD11a was successfully used in treatment of psoriasis vulgaris [266]. CD11a blockade using Efalizumab resulted in an unexpected downregulation of a broad range of T cell surface molecules including the TCR, costimulatory molecules and integrins unrelated to LFA-1, both in the peripheral circulation and in diseased skin. Unfortunately, Efalizumab had to be withdrawn from the market since it caused reactivation of the JC virus and subsequent progressive multifocal leukoencephalopathy in some patients [267,268].

5.2.2. MAC-1

In CD11b−/− mice, the onset of EAE was delayed, and the course of disease was less severe as compared to WT animals [269]. In accordance, CD4+ T cells derived from immunized CD11b−/− mice were characterized by elevated production of anti-inflammatory (e.g., TGF-β, IL-10) and attenuated generation of pro-inflammatory (e.g., IFN-γ, TNF-α) cytokines as compared to WT T cells. In agreement, adoptive transfer of encephalogeneic T cells from CD11b−/− mice to WT mice induced no EAE. Vice versa, transfer of encephalogeneic T cells from WT to CD11b−/− mice resulted in a mild course of disease. Taken together, these findings suggest that in EAE MAC-1 is required to induce a profound inflammatory response. Moreover, CD11b+ B cells were demonstrated to suppress TCR signaling in a mouse model of experimental autoimmune hepatitis [189].
In a murine collagen-induced arthritis (CIA) model, MAC-1 expression prevented early disease onset, and decreased the severity of CIA via controlling IL-6 secretion and subsequent Th17 polarization of lymphocytes [255]. On C57BL6 genetic background, CD11b−/− mice developed CIA of high severity and incidence, whereas WT mice were fully protected from the disease. [255]. Adoptive transfer of WT DC to CD11b−/− animals reduced the severity of arthritis. As mentioned above, MAC-1 controls in part the phagocytic activity of FcR [114]. In a humanized mouse model with leukocytes expressing human FcγRIIA, CD11b deficiency protected from lupus nephritis that normally develops in response to injection of human SLE sera [270]. The role of MAC-1 for PMN-mediated autoimmune diseases was assessed in a model of Fc-dependent anti-GBM (glomerular basement membrane) nephritis [271]. In this model, application of anti-GBM antibody results in glomerular PMN accumulation due to binding/uptake of GBM/antibody immune complexes via FcR. In CD11b−/− mice, PMN infiltrated glomeruli initially at similar extent as observed for WT animals, but later on at much lower rate [272]. In agreement, CD11b−/− mice developed no proteinuria, in contrast to WT mice. Ex vivo analysis of PMN showed that MAC-1 was not required for PMN migration and adhesion but that interaction of MAC-1 and FcR was necessary for for F-actin rearrangements leading to long-lasting PMN adhesion. Bullous pemphigoid (BP) is an IgG-mediated autoimmune disease resulting in skin blisters [273]. In mice, BP is induced by injection of the hemidesmosome antigen BP180 mediating accumulation of CD11b+ cells in the skin that confer inflammatory reactions [274]. In CD11b−/− mice the development of BP in mice was significantly lower as compared with WT mice, since CD11b−/− PMN could not efficiently infiltrate the skin [275].
In agreement with the role of MAC-1 to contribute to the induction and maintenance of tolerancein, human CD11b gene polymorphismus which inactivated CD11b were shown to be associated with development of SLE and RA [276]. Three common CD11b SNP (single-nucleotide polymorphisms) translate into miss-sense mutations (P1146S, R77H, A858V), which are associated with SLE pathogenesis [277] and its common complication, lupus nephritis [278].

5.2.3. Other β2 Integrins

Compared to LFA-1 and MAC-1, the role of CD11c/CD18 (CR4) and CD11d/CD18 for the induction and progression of autoimmune diseases has been studied less intensively. Concerning CD11c, it was shown that the iC3b binding site of CR4 was required for the induction of the delayed type hypersensitivity [279,280]. Aziz and co-workers reported that M1 macrophages and white adipose tissue upregulated the expression of the CD11d, which in turn may contribute to the chronification of inflammation [67]. Furthermore, it has been noted that antibody-mediated activation of CD11d/CD18 increased IL-1β expression [281], known as strongly associated with acute and chronic inflammation [282].
To sum up, all four β2 integrins contribute to inflammation. LFA-1 and MAC-1 have been extensively studied in various rodent autoimmune models like EAE, SLE, and CIA and contribute to (auto)inflammation by supporting cell migration, uptake of autoantigens, subsequent stimulatory cell signaling, and T cell activation.

5.3. β2 Integrins and Tumor Development

Whereas tumor infiltration by T cells that commonly express LFA-1 strongly correlates with overall prognosis and with response to checkpoint inhibitor-based immunotherapy in mice and men, tumor infiltration with myeloid cells is often associated with poor prognosis and rapid tumor growth [298]. In established solid tumors, tumor-infiltrating monocytes differentiate to tumor-associated macrophages (TAM) [299]. TAM primarily serve to promote tumor growth by various mechanisms, including the generation of angiogenetic factors like VEGF (vascular endothelial growth factor) and the release of immunomodulatory factors that counteract tumor-infiltrating immune effector cells [300]. Moreover, immunomodulatory cytokines as generated by TAM and tumor cells interfere with the differentiation of myeloid progenitor cells [301]. This results in the induction of monocytic (CD11b+Ly-6C+) and granulocytic (CD11b+Ly-6G+) myeloid-derived suppressor cells (MDSC) [302]. Similar to TAM, MDSC accumulate in the TME but also in lymphoid tissues exerting immunosuppressive functions by directly tolerizing APC and inhibiting effector T cells by various mechanisms [303]. In the following, the role of β2 integrins for tumor development are discussed, and major findings are summarized in Table 2.

5.3.1. Tumor Infiltration

Infiltration of CD18+ cells early in tumor development was reported to prevent its progression. [304]. However, at later stages of tumor development infiltration of a tumor by myeloid cells may turn into a disadvantage, since TAM and MDSC that can induce tolerance localize to the tumor site and support its progression by various mechanisms [305,306,307]. Immunohistochemical analysis of human gastric tumor tissues revealed that most tumor infiltrating CD11b+ cells were (conventional) CD11c+ DC and that high infiltration of these cells correlated with tumor size, venous invasion, lymph node metastasis, the general metastasis stage, and infiltration by FoxP3+ Treg [308]. Accordingly, patients with high CD11b+ cell infiltration of the tumor had a poor outcome [281,307,308]. In agreement, Zhang and colleagues showed that CD11b deficiency in mice resulted in reduced infiltration of spontaneous intestinal adenoma with myeloid cells and attenuated tumor growth [305]. Systemic application of CD11b-specific monoclonal antibodies increased the anti-tumor response after radiation in mice, as myeloid cells could not migrate to the tumor site and support tumor angiogenesis (see below) [309].

5.3.2. Tumor Angiogenesis

A study conveyed by Soloviev and colleagues has shown that not only migration but as well VEGF secretion is orchestrated by β2 integrins and influences tumor fate [310]. In this regard, CD11b−/− but not CD11a−/− mice inoculated with B16F10 melanoma or RM1 prostate cancer cells displayed attenuated tumor neovascularization as compared to WT mice. One reason for this is an impaired infiltration of tumor tissue with PMNs and macrophages, which secrete VEGF needed for vessel development. In addition, CD11b−/− PMNs were characterized by markedly reduced degranulation and VEGF secretion upon TNF-α stimulation [310,311].

5.3.3. Tumor-Specific Immune Responses

As discussed above, MAC-1/CR3 and CR4 confer uptake of complement-opsonized material, and MAC-1 is involved in FcR-mediated internalization of antibody-opsonized cells. As a consequence, anti-melanoma antibody treatment for induction of ADCC (antibody-dependent cytotoxicity) in a melanoma metastasis model [312] was much less effective in CD11b−/− mice as in WT animals [313]. Besides complement-opsonized material, MAC-1 (CR3) and, to lower extent, CD11c/CD18 (CR4) bind a variety of soluble ligands, many of which can be found within the TME (see Section 2.4). Interaction of tumor-associated ligands with β2 integrins and the resulting immunoregulation is not well defined yet. For example, apoptotic bodies derived from tumor cells are taken up via MAC-1 (in addition to CD36) by DC, resulting in tolerance induction [126]. Thus, endocytosis of tumor-derived material and the resulting immunological response are both regulated by β2 integrins.

5.3.4. Interaction with Tumor Cells

The adhesive properties of β2 integrins have been controversially described in the context of tumor growth control. In case of LFA-1, it has been shown that on one hand it is an essential modulator of the IS between an NK cell or CTL and a tumor cell and hence is responsible for both adhesion and targeted release of the cytotoxic granules that kill the tumor cell [314,315,316,317]. On the other hand, it has been reported that LFA-1 and MAC-1 mediate adhesion of PMN to ICAM-1-expressing melanoma cells, allowing tandem migration of tumor cells, and thereby their extravasation [318,319]. Thus, binding of a leukocyte via β2 integrins to a tumor cell may lead both to eradication of the tumor or to its metastasis.

5.3.5. Leukemia

In chronic lymphatic leukemia (CLL) overall CD18 expression by malignant leukocytes is lower in comparison to healthy cells [320], and a functionally impaired CD18 variant (E630K) has recently been described to correlate with disease susceptibility [321]. However, in the subgroup of Trisomy 12 (tri12) harboring CLL elevated expression of LFA-1 and hence transendothelial migration was reported by several groups [322,323]. Furthermore, in CLL the adhesion of malignant B cells via LFA-1 can be disturbed due to defective inside-out signaling, involving Rap-1 GTPase affecting both β2 integrins and VLA-4 [324]. This signaling defect resulted in retention of non-functional clonal mature B cells in the blood [325,326]. Again, the subgroup of tri12 CLL differed by displaying increased levels of integrin signaling adaptors like CALDAG–GEF I, Rap-1B (Rap-1 Binding Protein), and VLA-4 [322]. Whereas Rho GTPases was required for chemokine-induced LFA-1 triggering in all CLL patients, in some, the GTPases Rac1 and Cdc42 were found dispensable [327].

5.3.6. β2 Integrin Expression by Tumor Cells

Whereas LFA-1 expression is confined to leukocytes under normal conditions [6], tumor cells may express this β2 integrin as well, shown to elevate their metastatic activity [328]. The surface glykoprotein CD44 is frequently expressed by tumor cells and mediates tumor cell migration by binding collagen as a constituent of the ECM [329]. Cross-linking of CD44 on tumor cells was reported to induce LFA-1 and VLA-4 expression [330]. Both receptors equally mediate transendothelial tumor cell migration as deduced from the finding that antibody-mediated blockade of either receptor did not affect transendothelial tumor cell migration but only their combined inhibition.
In general, even though β2 integrins are required for any innate/adaptive immune response, tumor infiltration by leukocytes as mediated by LFA-1 and MAC-1 supports growth of an established tumor that already communicates with an environment in order to induce tolerance. A deeper understanding of the role of β2 integrins in the TME, especially with regard to their potential function in regulatory immune cells, may allow to develop new therapeutic strategies targeting specific β2 integrins in a cell type-specific manner.

6. β2 Integrins as Therapeutic Targets

In the last decades, β2 integrins have received much attention as therapeutic targets mostly for treatment of autoimmune conditions. Integrin antagonists, such as a humanized CD11a blocking antibody (Efalizumab), have been developed to inhibit LFA-1 activity and thereby to treat psoriasis [335]. However, Efalizumab was withdrawn from the market in 2009 due to JC virus reactivation in some patients [336,337]. This adverse effect was caused most likely by deficient leukocyte migration and subsequent immunodeficiency. Similar side effects, although less severe, were reported in patients treated with Natalizunamb, an antibody specific for α4 integrin [338,339]. This antibody binds α4β1 and α4β7 and has been successfully used to treat patients suffering from Morbus Crohn and MS [340,341]. The pharmacological drug BMS-587101 has been developed to selectively block CD11a and was intended to cause less severe side effects as monoclonal antibodies [342]. BMS-587101 was reported to effectively reduce lung inflammation and joint destruction in a murine RA model [343] and attenuated transplant rejection in a mouse model [342] but was not further developed. Currently, no active clinical trials with antibodies or antagonists to β2 Integrins are listed on clinicaltrials.gov.
Leukadherin-1 (LA-1), selectively activates MAC-1 and increases cell adhesion to ICAM [344]. It has been noted that LA-1 suppresses innate inflammatory signaling in human NK cells [130]. In contrast to LFA-1, As outlined above, MAC-1 not only mediates leukocyte migration, but as well contributes to tolerogenic signaling. NK cells pre-treated with LA-1 displayed less STAT5 phosphorylation in response to IL-12, and consequently reduced secretion of TNF-α and IFN-γ [130]. LA-1 has been successfully used to prevent inflammation in hypoxia-induced lung injury in rats [345] and in an autoimmune nephritis mouse model [346,347]. Since myeloid cells express both LFA-1 and MAC-1 that bind other (immune) cells via ICAM, specific blockade of either β2 integrin may be compensated by the other. Therefore, as an alternative approach, ICAM-1 blocking antibodies have been developed. A study on patients with early RA showed benefits of an ICAM-1 blocking antibody [348,349]. Unfortunately, side effects restricted further testing [348].

7. Concluding Remarks

β2 integrins strongly contribute to the functional activity of any type of immune cell analyzed so far, comprising regulation of innate immune functions as the recognition of pathogens, infiltration of inflamed tissue, and pathogen killing, as well as adaptive immune functions, including antigen uptake, and stimulation/polarization of T cells and B cells to induce pathogen-specific immune responses. However, triggering of β2 integrins on APC may also confer tolerance and may thus contribute to impaired clearance of pathogens and tumor development, which in turn promotes the induction/expansion of regulatory immune cells. Regarding the latter, intriguingly, the role of β2 integrins for the immunosuppressive activity and the crosstalk of regulatory immune cells, namely Treg, MDSC, and TAM, has scarcely been addressed yet. However, this knowledge is crucial to develop tailored intervention strategies that counteract the immune-inhibitory activity of regulatory immune cells.
By now, the pathophysiological role of ß2 integrins has been studied largely in various mouse models with a constitutive knock-out of either an individual α or the common β subunit. However, in all of these mouse models it is difficult to delineate at the cellular level in vivo whether any functional alteration is the consequence of an intrinsic defect or a result of crosstalk with other immune cells deficient for β2 integrins as well. To overcome these limits, mouse models that enable cell type-specific deletion of β2 integrins need to be established.
So far, β2 integrin-focused attempts to treat (autoimmune) diseases aimed to modulate the activation state of a given β2 integrin systemically. However, β2 integrins may exert contrary functions in a cell type-specific manner, e.g., limit T cell activation when expressed on APC (LFA-1, MAC-1), but be necessary for T cell activation (LFA-1) when expressed on T cells. Hence, multi-functionalized nano-therapeutics that co-deliver a cell type-targeting and a β2 integrin-modulating moiety may provide a suitable mean to overcome this obstacle.

Funding

This work was supported by the Deutsche Forschungsgemeinschaft (TR156, project B11).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAPFyn-binding protein
ADCCAntibody-dependent cytotoxicity
ADMIDASAdjacent to metal-ion-dependent-adhesion-site
AP-1Activator protein-1
APCAntigen presenting cell
Arp2/3actin-related protein-2/3
B cellsB lymphocytes
BCGBacillus Calmette–Guérin
BCL6B cell lymphoma 6
BM-DCBone marrow-derived dendritic cells
BPBullous pemphigoid
Ca2Calcium
CALDAG–GEF ICa2+ and diacylglycerol regulated guanine nucleotide exchange factor I
Calf-1Calcium channel localization factor-1
Cas-LCrk-associated substrate lymphocyte-type
CDCluster of differentiation
CD40LCD40 ligand
CDC42Cell division control protein 42 homolog
cSMACcentral supramolecular activation cluster
CIACollagen-induced arthritis
CLLChronic lymphatic leukemia
CMVCytomegalovirus
CTLcytotoxic T lymphocytes
Cyr61Cysteine-rich angiogenic inducer 61
CYTIPCytohesin-1 interacting protein
DCDendritic cells
DC-SIGNDendritic cell-specific ICAM-3-grabbing non-integrin
DENND1CDifferentially expressed in normal and neoplastic cells domain 1C
DOCK2Dedicator of cytokinesis 2
EAEExperimental autoimmune encephalomyelitis
EGFEpidermal growth factor
EOCEpithelial ovarian cancer
ERKExtracellular signal-regulated kinase
ESEmbryonic stem cells
FcRFc receptor
FcγRIIAConventional type I transmembrane protein
FERMT3Fermitin family homolog 3
Foxp3Forkhead-Box-Protein P3
GEFGuanine nucleotide exchange factor
hHuman
HSPCHematopoietic stem and progenitor cells
ICAMIntercellular adhesion molecule
IFN-yInterferon y
IL-6Interleukin-6
ISimmunological synapse
ITGAMIntegrin subunit alpha M
JAB1Jun activating binding protein-1
JAKJanus kinase
JAMJunctional adhesion molecule
LA-1Leukadherin-1
LAD-ILeukocyte adhesion deficiency type 1
LckLymphocyte cell-specific protein tyrosine kinase
LIMBSLigand-associated metal binding site
LLCLewis lung carcinoma
LPSLipopolysaccharide
MA-ARDSMalaria-associated acute respiratory distress syndrome
MAC-1Macrophage antigen 1
MAPKMitogen activated protein kinase
MHCmajor histocompatibility complex
MIDASMetal ion-dependent-adhesion-site
MIPMacrophage inflammatory protein
MOGMyelin oligodendrocyte glycoprotein
mMouse
MPOMyeloperoxidase
MSMultiple sclerosis
Mst1Macrophage-Stimulating Protein
NFATc1nuclear factor of activated T cells, cytoplasmic 1
NK cellsNatural killer cells
NOTCH1Neurogenic locus notch homolog protein 1
NOX2NADPH Oxidase 2
PI3KPhosphoinositide 3-kinase
PIP5K1CPhosphatidylinositol-4-phosphate 5-kinase type-1 gamma
PKCProtein kinase C
PLCPhospholipase C
PLD1Phospholipase D1
PMNPolymorphonuclear granulocytes
PRLPhosphatase of regenerating liver 1
PSIPlexin-semaphorin-integrin
PSGL-1P-selectin glycoprotein ligand-1
pSMACPeripheral supramolecular activation cluster
PTPRGProtein tyrosine phosphatase receptor type g
RARheumatoid arthritis
Rac1Ras-related C3 botulinum toxin substrate 1
RACK1Receptor for activated C-kinase 1
RAGEReceptor for advanced glycation end products
RANKLReceptor activator of nuclear factor kappa-Β ligand
Rap-1Ras-related protein 1
Rap1BRap-1 binding protein
RAPLRegulator of adhesion and polarization enriched in lymphocytes
RhoARas homolog gene family, member A
RIAMRap-1-GTP interacting adaptor molecule
ROSReactive oxygen species
SHP-1Src homology region 2 domain-containing phosphatase 1
SKAP55,Src kinase-associated phosphoprotein 5
SLESystemic lupus erythematosus
SNPSingle Nucleotide Polymorphism
SOCS-3Suppressor of cytokine signaling 3
SPTAN1Spectrin alpha, non-erythrocytic 1
STATSignal transducers and activators of transcription
STK4Serine/threonine protein kinase 4
SYKSpleen tyrosine kinase
T cellsT lymphocytes
TCRT cell receptor
TEMTransmission electron microscopy
TGLN2Taglin 2
Thy-1Thymus cell antigen 1
TLRToll-like Receptor
TMETumor microenvironment
TNF-αTumor necrosis factor alpha
TregRegulatory T cells
Tri12Trisomy 12
VAV1Vav Guanine Nucleotide Exchange Factor 1
VCAM-1Vascular cell adhesion protein 1
VEGFVascular endothelial growth factor
WASpWiskott-Aldrich syndrome protein
WBCWhite blood cells
WTWild type
γ/δ T cellsGamma/delta T cells

References

  1. Takada, Y.; Ye, X.; Simon, S. The integrins. Genome Biol. 2007, 8, 215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Johnson, M.S.; Lu, N.; Denessiouk, K.; Heino, J.; Gullberg, D. Integrins during evolution: Evolutionary trees and model organisms. Biochim. Et Biophys. Acta 2009, 1788, 779–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. LaFoya, B.; Munroe, J.A.; Miyamoto, A.; Detweiler, M.A.; Crow, J.J.; Gazdik, T.; Albig, A.R. Beyond the Matrix: The Many Non-ECM Ligands for Integrins. Int. J. Mol. Sci. 2018, 19, 449. [Google Scholar] [CrossRef] [Green Version]
  4. Conway, J.R.W.; Jacquemet, G. Cell matrix adhesion in cell migration. Essays Biochem. 2019, 63, 535–551. [Google Scholar] [CrossRef] [PubMed]
  5. Bros, M.; Haas, K.; Moll, L.; Grabbe, S. RhoA as a Key Regulator of Innate and Adaptive Immunity. Cells 2019, 8, 733. [Google Scholar] [CrossRef] [Green Version]
  6. Mitroulis, I.; Alexaki, V.I.; Kourtzelis, I.; Ziogas, A.; Hajishengallis, G.; Chavakis, T. Leukocyte integrins: Role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol. Ther. 2015, 147, 123–135. [Google Scholar] [CrossRef] [Green Version]
  7. Cabanillas, D.; Regairaz, L.; Deswarte, C.; Garcia, M.; Richard, M.E.; Casanova, J.L.; Bustamante, J.; Perez, L. Leukocyte Adhesion Deficiency Type 1 (LAD1) with Expressed but Nonfunctional CD11/CD18. J. Clin. Immunol 2016, 36, 627–630. [Google Scholar] [CrossRef]
  8. LaFlamme, S.E.; Mathew-Steiner, S.; Singh, N.; Colello-Borges, D.; Nieves, B. Integrin and microtubule crosstalk in the regulation of cellular processes. Cell. Mol. Life Sci. 2018, 75, 4177–4185. [Google Scholar] [CrossRef]
  9. Streuli, C.H.; Akhtar, N. Signal co-operation between integrins and other receptor systems. Biochem. J. 2009, 418, 491–506. [Google Scholar] [CrossRef] [Green Version]
  10. Humphries, M.J.; Symonds, E.J.; Mould, A.P. Mapping functional residues onto integrin crystal structures. Curr. Opin. Struct. Biol. 2003, 13, 236–243. [Google Scholar] [CrossRef]
  11. Chouhan, B.S.; Kapyla, J.; Denessiouk, K.; Denesyuk, A.; Heino, J.; Johnson, M.S. Early chordate origin of the vertebrate integrin alphaI domains. PLoS ONE 2014, 9, e112064. [Google Scholar] [CrossRef] [Green Version]
  12. Nunes, A.M.; Minetti, C.; Remeta, D.P.; Baum, J. Magnesium Activates Microsecond Dynamics to Regulate Integrin-Collagen Recognition. Structure 2018, 26, 1080–1090 e1085. [Google Scholar] [CrossRef] [Green Version]
  13. Sen, M.; Yuki, K.; Springer, T.A. An internal ligand-bound, metastable state of a leukocyte integrin, alphaXbeta2. J. Cell Biol. 2013, 203, 629–642. [Google Scholar] [CrossRef] [PubMed]
  14. Shimaoka, M.; Salas, A.; Yang, W.; Weitz-Schmidt, G.; Springer, T.A. Small molecule integrin antagonists that bind to the beta2 subunit I-like domain and activate signals in one direction and block them in the other. Immunity 2003, 19, 391–402. [Google Scholar] [CrossRef] [Green Version]
  15. Xu, S.; Wang, J.; Wang, J.H.; Springer, T.A. Distinct recognition of complement iC3b by integrins alphaXbeta2 and alphaMbeta2. Proc. Natl. Acad. Sci. USA 2017, 114, 3403–3408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Adorno-Cruz, V.; Liu, H. Regulation and functions of integrin alpha2 in cell adhesion and disease. Genes Dis. 2019, 6, 16–24. [Google Scholar] [CrossRef]
  17. Lim, J.; Hotchin, N.A. Signalling mechanisms of the leukocyte integrin alphaMbeta2: Current and future perspectives. Biol. Cell 2012, 104, 631–640. [Google Scholar] [CrossRef]
  18. Zhang, K.; Chen, J. The regulation of integrin function by divalent cations. Cell Adh. Migr. 2012, 6, 20–29. [Google Scholar] [CrossRef] [Green Version]
  19. Xiang, B.; Liu, Y.; Xie, L.; Zhao, Q.; Zhang, L.; Gan, X.; Yu, H. The osteoclasts attach to the bone surface where the extracellular calcium concentration decreases. Cell Biochem. Biophys. 2016, 74, 553–558. [Google Scholar] [CrossRef]
  20. Valdramidou, D.; Humphries, M.J.; Mould, A.P. Distinct roles of beta1 metal ion-dependent adhesion site (MIDAS), adjacent to MIDAS (ADMIDAS), and ligand-associated metal-binding site (LIMBS) cation-binding sites in ligand recognition by integrin alpha2beta1. J. Biol. Chem. 2008, 283, 32704–32714. [Google Scholar] [CrossRef] [Green Version]
  21. Rui, X.; Mehrbod, M.; Van Agthoven, J.F.; Anand, S.; Xiong, J.P.; Mofrad, M.R.; Arnaout, M.A. The alpha-subunit regulates stability of the metal ion at the ligand-associated metal ion-binding site in beta3 integrins. J. Biol. Chem. 2014, 289, 23256–23263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Thome, S.; Begandt, D.; Pick, R.; Salvermoser, M.; Walzog, B. Intracellular beta2 integrin (CD11/CD18) interacting partners in neutrophil trafficking. Eur. J. Clin. Investig. 2018, 48 (Suppl. 2), e12966. [Google Scholar] [CrossRef] [Green Version]
  23. Muller, W.A. Getting leukocytes to the site of inflammation. Vet. Pathol 2013, 50, 7–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Varga, G.; Balkow, S.; Wild, M.K.; Stadtbaeumer, A.; Krummen, M.; Rothoeft, T.; Higuchi, T.; Beissert, S.; Wethmar, K.; Scharffetter-Kochanek, K.; et al. Active MAC-1 (CD11b/CD18) on DCs inhibits full T-cell activation. Blood 2007, 109, 661–669. [Google Scholar] [CrossRef] [PubMed]
  25. Jung, S.; Yuki, K. Differential effects of volatile anesthetics on leukocyte integrin macrophage-1 antigen. J. Immunotoxicol. 2016, 13, 148–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Block, H.; Stadtmann, A.; Riad, D.; Rossaint, J.; Sohlbach, C.; Germena, G.; Wu, D.; Simon, S.I.; Ley, K.; Zarbock, A. Gnb isoforms control a signaling pathway comprising Rac1, Plcbeta2, and Plcbeta3 leading to LFA-1 activation and neutrophil arrest in vivo. Blood 2016, 127, 314–324. [Google Scholar] [CrossRef] [Green Version]
  27. Jaskiewicz, A.; Pajak, B.; Orzechowski, A. The Many Faces of Rap1 GTPase. Int. J. Mol. Sci. 2018, 19, 2848. [Google Scholar] [CrossRef] [Green Version]
  28. Hashimoto, A.; Tanaka, M.; Takeda, S.; Ito, H.; Nagano, K. Cilostazol Induces PGI2 Production via Activation of the Downstream Epac-1/Rap1 Signaling Cascade to Increase Intracellular Calcium by PLCepsilon and to Activate p44/42 MAPK in Human Aortic Endothelial Cells. PLoS ONE 2015, 10, e0132835. [Google Scholar] [CrossRef]
  29. Katagiri, K.; Maeda, A.; Shimonaka, M.; Kinashi, T. RAPL, a Rap1-binding molecule that mediates Rap1-induced adhesion through spatial regulation of LFA-1. Nat. Immunol. 2003, 4, 741–748. [Google Scholar] [CrossRef]
  30. Shannon, M.J.; Pineau, J.; Griffie, J.; Aaron, J.; Peel, T.; Williamson, D.J.; Zamoyska, R.; Cope, A.P.; Cornish, G.H.; Owen, D.M. Differential nanoscale organisation of LFA-1 modulates T cell migration. J. Cell Sci. 2019. [Google Scholar] [CrossRef] [Green Version]
  31. Witte, A.; Meineke, B.; Sticht, J.; Philipsen, L.; Kuropka, B.; Muller, A.J.; Freund, C.; Schraven, B.; Kliche, S. D120 and K152 within the PH Domain of T Cell Adapter SKAP55 Regulate Plasma Membrane Targeting of SKAP55 and LFA-1 Affinity Modulation in Human T Lymphocytes. Mol. Cell. Biol. 2017, 37. [Google Scholar] [CrossRef] [Green Version]
  32. Stadtmann, A.; Zarbock, A. The role of kindlin in neutrophil recruitment to inflammatory sites. Curr. Opin. Hematol. 2017, 24, 38–45. [Google Scholar] [CrossRef] [PubMed]
  33. Wegener, K.L.; Partridge, A.W.; Han, J.; Pickford, A.R.; Liddington, R.C.; Ginsberg, M.H.; Campbell, I.D. Structural basis of integrin activation by talin. Cell 2007, 128, 171–182. [Google Scholar] [CrossRef] [PubMed]
  34. Patsoukis, N.; Bardhan, K.; Weaver, J.D.; Sari, D.; Torres-Gomez, A.; Li, L.; Strauss, L.; Lafuente, E.M.; Boussiotis, V.A. The adaptor molecule RIAM integrates signaling events critical for integrin-mediated control of immune function and cancer progression. Sci. Signal. 2017, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Fan, Z.; Ley, K. Leukocyte arrest: Biomechanics and molecular mechanisms of beta2 integrin activation. Biorheology 2015, 52, 353–377. [Google Scholar] [CrossRef] [Green Version]
  36. Lefort, C.T.; Rossaint, J.; Moser, M.; Petrich, B.G.; Zarbock, A.; Monkley, S.J.; Critchley, D.R.; Ginsberg, M.H.; Fassler, R.; Ley, K. Distinct roles for talin-1 and kindlin-3 in LFA-1 extension and affinity regulation. Blood 2012, 119, 4275–4282. [Google Scholar] [CrossRef]
  37. Yago, T.; Zhang, N.; Zhao, L.; Abrams, C.S.; McEver, R.P. Selectins and chemokines use shared and distinct signals to activate beta2 integrins in neutrophils. Blood Adv. 2018, 2, 731–744. [Google Scholar] [CrossRef] [Green Version]
  38. Geiger, C.; Nagel, W.; Boehm, T.; van Kooyk, Y.; Figdor, C.G.; Kremmer, E.; Hogg, N.; Zeitlmann, L.; Dierks, H.; Weber, K.S.; et al. Cytohesin-1 regulates beta-2 integrin-mediated adhesion through both ARF-GEF function and interaction with LFA-1. Embo J. 2000, 19, 2525–2536. [Google Scholar] [CrossRef] [Green Version]
  39. Quast, T.; Tappertzhofen, B.; Schild, C.; Grell, J.; Czeloth, N.; Forster, R.; Alon, R.; Fraemohs, L.; Dreck, K.; Weber, C.; et al. Cytohesin-1 controls the activation of RhoA and modulates integrin-dependent adhesion and migration of dendritic cells. Blood 2009, 113, 5801–5810. [Google Scholar] [CrossRef] [Green Version]
  40. Rognoni, E.; Ruppert, R.; Fassler, R. The kindlin family: Functions, signaling properties and implications for human disease. J. Cell Sci. 2016, 129, 17–27. [Google Scholar] [CrossRef] [Green Version]
  41. Li, N.; Yang, H.; Wang, M.; Lu, S.; Zhang, Y.; Long, M. Ligand-specific binding forces of LFA-1 and Mac-1 in neutrophil adhesion and crawling. Mol. Biol. Cell 2018, 29, 408–418. [Google Scholar] [CrossRef] [PubMed]
  42. Wingren, A.G.; Parra, E.; Varga, M.; Kalland, T.; Sjogren, H.O.; Hedlund, G.; Dohlsten, M. T Cell Activation Pathways: B7, LFA-3, and ICAM-1 Shape Unique T Cell Profiles. Crit. Rev. Immunol. 2017, 37, 463–481. [Google Scholar] [CrossRef] [PubMed]
  43. Atsaves, V.; Leventaki, V.; Rassidakis, G.Z.; Claret, F.X. AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers (Basel) 2019, 11, 1037. [Google Scholar] [CrossRef] [Green Version]
  44. Pantarelli, C.; Welch, H.C.E. Rac-GTPases and Rac-GEFs in neutrophil adhesion, migration and recruitment. Eur. J. Clin. Investig. 2018, 48 (Suppl. 2), e12939. [Google Scholar] [CrossRef] [Green Version]
  45. Nurmi, S.M.; Autero, M.; Raunio, A.K.; Gahmberg, C.G.; Fagerholm, S.C. Phosphorylation of the LFA-1 integrin beta2-chain on Thr-758 leads to adhesion, Rac-1/Cdc42 activation, and stimulation of CD69 expression in human T cells. J. Biol. Chem. 2007, 282, 968–975. [Google Scholar] [CrossRef] [Green Version]
  46. Stadtmann, A.; Brinkhaus, L.; Mueller, H.; Rossaint, J.; Bolomini-Vittori, M.; Bergmeier, W.; Van Aken, H.; Wagner, D.D.; Laudanna, C.; Ley, K.; et al. Rap1a activation by CalDAG-GEFI and p38 MAPK is involved in E-selectin-dependent slow leukocyte rolling. Eur. J. Immunol. 2011, 41, 2074–2085. [Google Scholar] [CrossRef] [Green Version]
  47. Toffali, L.; Montresor, A.; Mirenda, M.; Scita, G.; Laudanna, C. SOS1, ARHGEF1, and DOCK2 rho-GEFs Mediate JAK-Dependent LFA-1 Activation by Chemokines. J. Immunol. 2017, 198, 708–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Riteau, B.; Barber, D.F.; Long, E.O. Vav1 phosphorylation is induced by beta2 integrin engagement on natural killer cells upstream of actin cytoskeleton and lipid raft reorganization. J. Exp. Med. 2003, 198, 469–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Burbach, B.J.; Srivastava, R.; Ingram, M.A.; Mitchell, J.S.; Shimizu, Y. The pleckstrin homology domain in the SKAP55 adapter protein defines the ability of the adapter protein ADAP to regulate integrin function and NF-kappaB activation. J. Immunol. 2011, 186, 6227–6237. [Google Scholar] [CrossRef] [Green Version]
  50. March, M.E.; Long, E.O. beta2 integrin induces TCRzeta-Syk-phospholipase C-gamma phosphorylation and paxillin-dependent granule polarization in human NK cells. J. Immunol. 2011, 186, 2998–3005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Xu, W.; Wang, P.; Petri, B.; Zhang, Y.; Tang, W.; Sun, L.; Kress, H.; Mann, T.; Shi, Y.; Kubes, P.; et al. Integrin-induced PIP5K1C kinase polarization regulates neutrophil polarization, directionality, and in vivo infiltration. Immunity 2010, 33, 340–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Montresor, A.; Toffali, L.; Constantin, G.; Laudanna, C. Chemokines and the signaling modules regulating integrin affinity. Front. Immunol. 2012, 3, 127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Urlaub, D.; Hofer, K.; Muller, M.L.; Watzl, C. LFA-1 Activation in NK Cells and Their Subsets: Influence of Receptors, Maturation, and Cytokine Stimulation. J. Immunol. 2017, 198, 1944–1951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kummer, D.; Ebnet, K. Junctional Adhesion Molecules (JAMs): The JAM-Integrin Connection. Cells 2018, 7, 25. [Google Scholar] [CrossRef] [Green Version]
  55. Podolnikova, N.P.; Podolnikov, A.V.; Haas, T.A.; Lishko, V.K.; Ugarova, T.P. Ligand recognition specificity of leukocyte integrin alphaMbeta2 (Mac-1, CD11b/CD18) and its functional consequences. Biochemistry 2015, 54, 1408–1420. [Google Scholar] [CrossRef] [Green Version]
  56. Lukacsi, S.; Nagy-Balo, Z.; Erdei, A.; Sandor, N.; Bajtay, Z. The role of CR3 (CD11b/CD18) and CR4 (CD11c/CD18) in complement-mediated phagocytosis and podosome formation by human phagocytes. Immunol. Lett. 2017, 189, 64–72. [Google Scholar] [CrossRef] [Green Version]
  57. Vorup-Jensen, T.; Jensen, R.K. Structural Immunology of Complement Receptors 3 and 4. Front. Immunol. 2018, 9, 2716. [Google Scholar] [CrossRef] [Green Version]
  58. Jacobson, A.C.; Weis, J.H. Comparative functional evolution of human and mouse CR1 and CR2. J. Immunol. 2008, 181, 2953–2959. [Google Scholar] [CrossRef]
  59. Humphries, J.D.; Byron, A.; Humphries, M.J. Integrin ligands at a glance. J. Cell Sci. 2006, 119, 3901–3903. [Google Scholar] [CrossRef] [Green Version]
  60. Yakubenko, V.P.; Yadav, S.P.; Ugarova, T.P. Integrin alphaDbeta2, an adhesion receptor up-regulated on macrophage foam cells, exhibits multiligand-binding properties. Blood 2006, 107, 1643–1650. [Google Scholar] [CrossRef]
  61. Yakubenko, V.P.; Cui, K.; Ardell, C.L.; Brown, K.E.; West, X.Z.; Gao, D.; Stefl, S.; Salomon, R.G.; Podrez, E.A.; Byzova, T.V. Oxidative modifications of extracellular matrix promote the second wave of inflammation via beta2 integrins. Blood 2018, 132, 78–88. [Google Scholar] [CrossRef] [PubMed]
  62. Walling, B.L.; Kim, M. LFA-1 in T Cell Migration and Differentiation. Front. Immunol. 2018, 9, 952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lim, K.; Hyun, Y.M.; Lambert-Emo, K.; Topham, D.J.; Kim, M. Visualization of integrin Mac-1 in vivo. J. Immunol. Methods 2015, 426, 120–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lishko, V.K.; Yakubenko, V.P.; Ugarova, T.P.; Podolnikova, N.P. Leukocyte integrin Mac-1 (CD11b/CD18, alphaMbeta2, CR3) acts as a functional receptor for platelet factor 4. J. Biol. Chem. 2018, 293, 6869–6882. [Google Scholar] [CrossRef] [Green Version]
  65. Singh-Jasuja, H.; Thiolat, A.; Ribon, M.; Boissier, M.C.; Bessis, N.; Rammensee, H.G.; Decker, P. The mouse dendritic cell marker CD11c is down-regulated upon cell activation through Toll-like receptor triggering. Immunobiology 2013, 218, 28–39. [Google Scholar] [CrossRef]
  66. Karnell, J.L.; Kumar, V.; Wang, J.; Wang, S.; Voynova, E.; Ettinger, R. Role of CD11c(+) T-bet(+) B cells in human health and disease. Cell. Immunol. 2017, 321, 40–45. [Google Scholar] [CrossRef]
  67. Aziz, M.H.; Cui, K.; Das, M.; Brown, K.E.; Ardell, C.L.; Febbraio, M.; Pluskota, E.; Han, J.; Wu, H.; Ballantyne, C.M.; et al. The Upregulation of Integrin alphaDbeta2 (CD11d/CD18) on Inflammatory Macrophages Promotes Macrophage Retention in Vascular Lesions and Development of Atherosclerosis. J. Immunol. 2017, 198, 4855–4867. [Google Scholar] [CrossRef] [Green Version]
  68. Yu, X.; Feng, B.; He, P.; Shan, L. From Chaos to Harmony: Responses and Signaling upon Microbial Pattern Recognition. Annu. Rev. Phytopathol. 2017, 55, 109–137. [Google Scholar] [CrossRef]
  69. Todd, J.L.; Palmer, S.M. Danger signals in regulating the immune response to solid organ transplantation. J. Clin. Investig. 2017, 127, 2464–2472. [Google Scholar] [CrossRef] [Green Version]
  70. Bose, T.O.; Colpitts, S.L.; Pham, Q.M.; Puddington, L.; Lefrancois, L. CD11a is essential for normal development of hematopoietic intermediates. J. Immunol. 2014, 193, 2863–2872. [Google Scholar] [CrossRef] [Green Version]
  71. Singh, K.; Gatzka, M.; Peters, T.; Borkner, L.; Hainzl, A.; Wang, H.; Sindrilaru, A.; Scharffetter-Kochanek, K. Reduced CD18 levels drive regulatory T cell conversion into Th17 cells in the CD18hypo PL/J mouse model of psoriasis. J. Immunol. 2013, 190, 2544–2553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Marski, M.; Kandula, S.; Turner, J.R.; Abraham, C. CD18 is required for optimal development and function of CD4+CD25+ T regulatory cells. J. Immunol. 2005, 175, 7889–7897. [Google Scholar] [CrossRef] [Green Version]
  73. Meli, A.P.; Fontes, G.; Avery, D.T.; Leddon, S.A.; Tam, M.; Elliot, M.; Ballesteros-Tato, A.; Miller, J.; Stevenson, M.M.; Fowell, D.J.; et al. The Integrin LFA-1 Controls T Follicular Helper Cell Generation and Maintenance. Immunity 2016, 45, 831–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Jogdand, G.M.; Mohanty, S.; Devadas, S. Regulators of Tfh Cell Differentiation. Front. Immunol. 2016, 7, 520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Vinuesa, C.G.; Linterman, M.A.; Yu, D.; MacLennan, I.C. Follicular Helper T Cells. Annu. Rev. Immunol. 2016, 34, 335–368. [Google Scholar] [CrossRef]
  76. Chen, X.; Wang, Z.; Duan, N.; Zhu, G.; Schwarz, E.M.; Xie, C. Osteoblast-osteoclast interactions. Connect. Tissue Res. 2018, 59, 99–107. [Google Scholar] [CrossRef]
  77. Lampiasi, N.; Russo, R.; Zito, F. The Alternative Faces of Macrophage Generate Osteoclasts. Biomed. Res. Int. 2016, 2016, 9089610. [Google Scholar] [CrossRef] [Green Version]
  78. Park-Min, K.H.; Lee, E.Y.; Moskowitz, N.K.; Lim, E.; Lee, S.K.; Lorenzo, J.A.; Huang, C.; Melnick, A.M.; Purdue, P.E.; Goldring, S.R.; et al. Negative regulation of osteoclast precursor differentiation by CD11b and beta2 integrin-B-cell lymphoma 6 signaling. J. Bone Miner. Res. 2013, 28, 135–149. [Google Scholar] [CrossRef] [Green Version]
  79. Wu, H.; Rodgers, J.R.; Perrard, X.Y.; Perrard, J.L.; Prince, J.E.; Abe, Y.; Davis, B.K.; Dietsch, G.; Smith, C.W.; Ballantyne, C.M. Deficiency of CD11b or CD11d results in reduced staphylococcal enterotoxin-induced T cell response and T cell phenotypic changes. J. Immunol. 2004, 173, 297–306. [Google Scholar] [CrossRef] [Green Version]
  80. Meng, D.; Qin, Y.; Lu, N.; Fang, K.; Hu, Y.; Tian, Z.; Zhang, C. Kupffer Cells Promote the Differentiation of Adult Liver Hematopoietic Stem and Progenitor Cells into Lymphocytes via ICAM-1 and LFA-1 Interaction. Stem Cells Int. 2019, 2019, 4848279. [Google Scholar] [CrossRef]
  81. Krenkel, O.; Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 2017, 17, 306–321. [Google Scholar] [CrossRef] [PubMed]
  82. Crozat, K.; Eidenschenk, C.; Jaeger, B.N.; Krebs, P.; Guia, S.; Beutler, B.; Vivier, E.; Ugolini, S. Impact of beta2 integrin deficiency on mouse natural killer cell development and function. Blood 2011, 117, 2874–2882. [Google Scholar] [CrossRef] [PubMed]
  83. Kourtzelis, I.; Mitroulis, I.; von Renesse, J.; Hajishengallis, G.; Chavakis, T. From leukocyte recruitment to resolution of inflammation: The cardinal role of integrins. J. Leukoc. Biol. 2017, 102, 677–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Alon, R.; Feigelson, S.W. Chemokine-triggered leukocyte arrest: Force-regulated bi-directional integrin activation in quantal adhesive contacts. Curr. Opin. Cell Biol. 2012, 24, 670–676. [Google Scholar] [CrossRef]
  85. Heit, B.; Colarusso, P.; Kubes, P. Fundamentally different roles for LFA-1, Mac-1 and alpha4-integrin in neutrophil chemotaxis. J. Cell Sci. 2005, 118, 5205–5220. [Google Scholar] [CrossRef] [Green Version]
  86. Ding, Z.M.; Babensee, J.E.; Simon, S.I.; Lu, H.; Perrard, J.L.; Bullard, D.C.; Dai, X.Y.; Bromley, S.K.; Dustin, M.L.; Entman, M.L.; et al. Relative contribution of LFA-1 and Mac-1 to neutrophil adhesion and migration. J. Immunol. 1999, 163, 5029–5038. [Google Scholar]
  87. Dunne, J.L.; Ballantyne, C.M.; Beaudet, A.L.; Ley, K. Control of leukocyte rolling velocity in TNF-alpha-induced inflammation by LFA-1 and Mac-1. Blood 2002, 99, 336–341. [Google Scholar] [CrossRef]
  88. Sumagin, R.; Prizant, H.; Lomakina, E.; Waugh, R.E.; Sarelius, I.H. LFA-1 and Mac-1 define characteristically different intralumenal crawling and emigration patterns for monocytes and neutrophils in situ. J. Immunol. 2010, 185, 7057–7066. [Google Scholar] [CrossRef] [Green Version]
  89. Trus, E.; Basta, S.; Gee, K. Who’s in charge here? Macrophage colony stimulating factor and granulocyte macrophage colony stimulating factor: Competing factors in macrophage polarization. Cytokine 2019, 127, 154939. [Google Scholar] [CrossRef]
  90. Cui, K.; Ardell, C.L.; Podolnikova, N.P.; Yakubenko, V.P. Distinct Migratory Properties of M1, M2, and Resident Macrophages Are Regulated by alphaDbeta2 and alphaMbeta2 Integrin-Mediated Adhesion. Front. Immunol. 2018, 9, 2650. [Google Scholar] [CrossRef]
  91. Wethmar, K.; Helmus, Y.; Luhn, K.; Jones, C.; Laskowska, A.; Varga, G.; Grabbe, S.; Lyck, R.; Engelhardt, B.; Bixel, M.G.; et al. Migration of immature mouse DC across resting endothelium is mediated by ICAM-2 but independent of beta2-integrins and murine DC-SIGN homologues. Eur. J. Immunol. 2006, 36, 2781–2794. [Google Scholar] [CrossRef] [PubMed]
  92. Heufler, C.; Ortner, D.; Hofer, S. Cybr, CYTIP or CASP: An attempt to pinpoint a molecule’s functions and names. Immunobiology 2008, 213, 729–732. [Google Scholar] [CrossRef] [PubMed]
  93. Balkow, S.; Heinz, S.; Schmidbauer, P.; Kolanus, W.; Holzmann, B.; Grabbe, S.; Laschinger, M. LFA-1 activity state on dendritic cells regulates contact duration with T cells and promotes T-cell priming. Blood 2010, 116, 1885–1894. [Google Scholar] [CrossRef] [PubMed]
  94. Theodoridis, A.A.; Eich, C.; Figdor, C.G.; Steinkasserer, A. Infection of dendritic cells with herpes simplex virus type 1 induces rapid degradation of CYTIP, thereby modulating adhesion and migration. Blood 2011, 118, 107–115. [Google Scholar] [CrossRef] [PubMed]
  95. Grosche, L.; Drassner, C.; Muhl-Zurbes, P.; Kamm, L.; Le-Trilling, V.T.K.; Trilling, M.; Steinkasserer, A.; Heilingloh, C.S. Human Cytomegalovirus-Induced Degradation of CYTIP Modulates Dendritic Cell Adhesion and Migration. Front. Immunol. 2017, 8, 461. [Google Scholar] [CrossRef] [PubMed]
  96. Morrison, V.L.; James, M.J.; Grzes, K.; Cook, P.; Glass, D.G.; Savinko, T.; Lek, H.S.; Gawden-Bone, C.; Watts, C.; Millington, O.R.; et al. Loss of beta2-integrin-mediated cytoskeletal linkage reprogrammes dendritic cells to a mature migratory phenotype. Nat. Commun. 2014, 5, 5359. [Google Scholar] [CrossRef] [Green Version]
  97. Savinko, T.S.; Morrison, V.L.; Uotila, L.M.; Wolff, C.H.J.; Alenius, H.T.; Fagerholm, S.C. Functional Beta2-Integrins Restrict Skin Inflammation In Vivo. J. Investig. Dermatol. 2015, 135, 2249–2257. [Google Scholar] [CrossRef] [Green Version]
  98. Nishikimi, A.; Ishihara, S.; Ozawa, M.; Etoh, K.; Fukuda, M.; Kinashi, T.; Katagiri, K. Rab13 acts downstream of the kinase Mst1 to deliver the integrin LFA-1 to the cell surface for lymphocyte trafficking. Sci. Signal. 2014, 7, ra72. [Google Scholar] [CrossRef]
  99. Xu, X.; Jaeger, E.R.; Wang, X.; Lagler-Ferrez, E.; Batalov, S.; Mathis, N.L.; Wiltshire, T.; Walker, J.R.; Cooke, M.P.; Sauer, K.; et al. Mst1 directs Myosin IIa partitioning of low and higher affinity integrins during T cell migration. PLoS ONE 2014, 9, e105561. [Google Scholar] [CrossRef]
  100. Samuelsson, M.; Potrzebowska, K.; Lehtonen, J.; Beech, J.P.; Skorova, E.; Uronen-Hansson, H.; Svensson, L. RhoB controls the Rab11-mediated recycling and surface reappearance of LFA-1 in migrating T lymphocytes. Sci. Signal. 2017, 10. [Google Scholar] [CrossRef] [Green Version]
  101. Takahashi, S.; Kubo, K.; Waguri, S.; Yabashi, A.; Shin, H.W.; Katoh, Y.; Nakayama, K. Rab11 regulates exocytosis of recycling vesicles at the plasma membrane. J. Cell Sci. 2012, 125, 4049–4057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Ong, S.T.; Freeley, M.; Skubis-Zegadlo, J.; Fazil, M.H.; Kelleher, D.; Fresser, F.; Baier, G.; Verma, N.K.; Long, A. Phosphorylation of Rab5a protein by protein kinase C is crucial for T-cell migration. J. Biol. Chem. 2014, 289, 19420–19434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Teske, C.; Schweitzer, C.; Palamidessi, A.; Aust, D.E.; Scita, G.; Weitz, J.; Welsch, T. Modulation of RAB5A early endosome trafficking in response to KRas mediated macropinocytic fluxes in pancreatic cancer cells. Biochem. Biophys. Res. Commun. 2017, 493, 528–533. [Google Scholar] [CrossRef] [PubMed]
  104. Rougerie, P.; Delon, J. Rho GTPases: Masters of T lymphocyte migration and activation. Immunol. Lett. 2012, 142, 1–13. [Google Scholar] [CrossRef]
  105. Jevnikar, Z.; Obermajer, N.; Pecar-Fonovic, U.; Karaoglanovic-Carmona, A.; Kos, J. Cathepsin X cleaves the beta2 cytoplasmic tail of LFA-1 inducing the intermediate affinity form of LFA-1 and alpha-actinin-1 binding. Eur. J. Immunol. 2009, 39, 3217–3227. [Google Scholar] [CrossRef]
  106. Rovira-Clave, X.; Angulo-Ibanez, M.; Reina, M.; Espel, E. The PDZ-binding domain of syndecan-2 inhibits LFA-1 high-affinity conformation. Cell. Signal. 2014, 26, 1489–1499. [Google Scholar] [CrossRef]
  107. Ueda, Y.; Kondo, N.; Ozawa, M.; Yasuda, K.; Tomiyama, T.; Kinashi, T. Sema3e/Plexin D1 Modulates Immunological Synapse and Migration of Thymocytes by Rap1 Inhibition. J. Immunol. 2016, 196, 3019–3031. [Google Scholar] [CrossRef] [Green Version]
  108. Mirenda, M.; Toffali, L.; Montresor, A.; Scardoni, G.; Sorio, C.; Laudanna, C. Protein tyrosine phosphatase receptor type gamma is a JAK phosphatase and negatively regulates leukocyte integrin activation. J. Immunol. 2015, 194, 2168–2179. [Google Scholar] [CrossRef] [Green Version]
  109. Chen, J.; Zhong, M.C.; Guo, H.; Davidson, D.; Mishel, S.; Lu, Y.; Rhee, I.; Perez-Quintero, L.A.; Zhang, S.; Cruz-Munoz, M.E.; et al. SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature 2017, 544, 493–497. [Google Scholar] [CrossRef] [Green Version]
  110. Hawley, K.L.; Olson, C.M., Jr.; Carreras-Gonzalez, A.; Navasa, N.; Anguita, J. Serum C3 Enhances Complement Receptor 3-Mediated Phagocytosis of Borrelia burgdorferi. Int. J. Biol. Sci. 2015, 11, 1269–1271. [Google Scholar] [CrossRef] [Green Version]
  111. Zhou, M.; Todd, R.F., 3rd; van de Winkel, J.G.; Petty, H.R. Cocapping of the leukoadhesin molecules complement receptor type 3 and lymphocyte function-associated antigen-1 with Fc gamma receptor III on human neutrophils. Possible role of lectin-like interactions. J. Immunol. 1993, 150, 3030–3041. [Google Scholar] [PubMed]
  112. Saggu, G.; Okubo, K.; Chen, Y.; Vattepu, R.; Tsuboi, N.; Rosetti, F.; Cullere, X.; Washburn, N.; Tahir, S.; Rosado, A.M.; et al. Cis interaction between sialylated FcgammaRIIA and the alphaI-domain of Mac-1 limits antibody-mediated neutrophil recruitment. Nat. Commun. 2018, 9, 5058. [Google Scholar] [CrossRef] [PubMed]
  113. Jongstra-Bilen, J.; Harrison, R.; Grinstein, S. Fcgamma-receptors induce Mac-1 (CD11b/CD18) mobilization and accumulation in the phagocytic cup for optimal phagocytosis. J. Biol. Chem. 2003, 278, 45720–45729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. van Spriel, A.B.; Leusen, J.H.; van Egmond, M.; Dijkman, H.B.; Assmann, K.J.; Mayadas, T.N.; van de Winkel, J.G. Mac-1 (CD11b/CD18) is essential for Fc receptor-mediated neutrophil cytotoxicity and immunologic synapse formation. Blood 2001, 97, 2478–2486. [Google Scholar] [CrossRef]
  115. Wu, Z.; Zhang, Z.; Lei, Z.; Lei, P. CD14: Biology and role in the pathogenesis of disease. Cytokine Growth Factor Rev. 2019, 48, 24–31. [Google Scholar] [CrossRef]
  116. Zanoni, I.; Ostuni, R.; Marek, L.R.; Barresi, S.; Barbalat, R.; Barton, G.M.; Granucci, F.; Kagan, J.C. CD14 controls the LPS-induced endocytosis of Toll-like receptor 4. Cell 2011, 147, 868–880. [Google Scholar] [CrossRef] [Green Version]
  117. Ling, G.S.; Bennett, J.; Woollard, K.J.; Szajna, M.; Fossati-Jimack, L.; Taylor, P.R.; Scott, D.; Franzoso, G.; Cook, H.T.; Botto, M. Integrin CD11b positively regulates TLR4-induced signalling pathways in dendritic cells but not in macrophages. Nat. Commun. 2014, 5, 3039. [Google Scholar] [CrossRef] [Green Version]
  118. Perera, P.Y.; Mayadas, T.N.; Takeuchi, O.; Akira, S.; Zaks-Zilberman, M.; Goyert, S.M.; Vogel, S.N. CD11b/CD18 acts in concert with CD14 and Toll-like receptor (TLR) 4 to elicit full lipopolysaccharide and taxol-inducible gene expression. J. Immunol. 2001, 166, 574–581. [Google Scholar] [CrossRef] [Green Version]
  119. Zhou, H.; Liao, J.; Aloor, J.; Nie, H.; Wilson, B.C.; Fessler, M.B.; Gao, H.M.; Hong, J.S. CD11b/CD18 (Mac-1) is a novel surface receptor for extracellular double-stranded RNA to mediate cellular inflammatory responses. J. Immunol. 2013, 190, 115–125. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, Q.; Lee, W.B.; Kang, J.S.; Kim, L.K.; Kim, Y.J. Integrin CD11b negatively regulates Mincle-induced signaling via the Lyn-SIRPalpha-SHP1 complex. Exp. Mol. Med. 2018, 50, e439. [Google Scholar] [CrossRef]
  121. Jiang, L.; Liu, G.; Ni, W.; Zhang, N.; Jie, J.; Xie, F.; Tai, G. The Combination of MBP and BCG-Induced Dendritic Cell Maturation through TLR2/TLR4 Promotes Th1 Activation In Vitro and Vivo. Mediat. Inflamm 2017, 2017, 1953680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Bai, Y.; Qian, C.; Qian, L.; Ma, F.; Hou, J.; Chen, Y.; Wang, Q.; Cao, X. Integrin CD11b negatively regulates TLR9-triggered dendritic cell cross-priming by upregulating microRNA-146a. J. Immunol. 2012, 188, 5293–5302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Yee, N.K.; Hamerman, J.A. beta(2) integrins inhibit TLR responses by regulating NF-kappaB pathway and p38 MAPK activation. Eur. J. Immunol. 2013, 43, 779–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Boonyatecha, N.; Sangphech, N.; Wongchana, W.; Kueanjinda, P.; Palaga, T. Involvement of Notch signaling pathway in regulating IL-12 expression via c-Rel in activated macrophages. Mol. Immunol. 2012, 51, 255–262. [Google Scholar] [CrossRef] [Green Version]
  125. Morelli, A.E.; Larregina, A.T.; Shufesky, W.J.; Zahorchak, A.F.; Logar, A.J.; Papworth, G.D.; Wang, Z.; Watkins, S.C.; Falo, L.D., Jr.; Thomson, A.W. Internalization of circulating apoptotic cells by splenic marginal zone dendritic cells: Dependence on complement receptors and effect on cytokine production. Blood 2003, 101, 611–620. [Google Scholar] [CrossRef]
  126. Skoberne, M.; Somersan, S.; Almodovar, W.; Truong, T.; Petrova, K.; Henson, P.M.; Bhardwaj, N. The apoptotic-cell receptor CR3, but not alphavbeta5, is a regulator of human dendritic-cell immunostimulatory function. Blood 2006, 108, 947–955. [Google Scholar] [CrossRef] [Green Version]
  127. Ding, C.; Ma, Y.; Chen, X.; Liu, M.; Cai, Y.; Hu, X.; Xiang, D.; Nath, S.; Zhang, H.G.; Ye, H.; et al. Integrin CD11b negatively regulates BCR signalling to maintain autoreactive B cell tolerance. Nat. Commun. 2013, 4, 2813. [Google Scholar] [CrossRef] [Green Version]
  128. Zhang, M.; Han, Y.; Han, C.; Xu, S.; Bao, Y.; Chen, Z.; Gu, Y.; Xia, D.; Cao, X. The beta2 integrin CD11b attenuates polyinosinic:polycytidylic acid-induced hepatitis by negatively regulating natural killer cell functions. Hepatol. (Baltim. Md.) 2009, 50, 1606–1616. [Google Scholar] [CrossRef]
  129. Wooten, D.K.; Xie, X.; Bartos, D.; Busche, R.A.; Longmore, G.D.; Watowich, S.S. Cytokine signaling through Stat3 activates integrins, promotes adhesion, and induces growth arrest in the myeloid cell line 32D. J. Biol. Chem. 2000, 275, 26566–26575. [Google Scholar] [CrossRef] [Green Version]
  130. Roberts, A.L.; Furnrohr, B.G.; Vyse, T.J.; Rhodes, B. The complement receptor 3 (CD11b/CD18) agonist Leukadherin-1 suppresses human innate inflammatory signalling. Clin. Exp. Immunol. 2016, 185, 361–371. [Google Scholar] [CrossRef] [Green Version]
  131. Basu, R.; Huse, M. Mechanical Communication at the Immunological Synapse. Trends Cell Biol. 2017, 27, 241–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Benvenuti, F. The Dendritic Cell Synapse: A Life Dedicated to T Cell Activation. Front. Immunol. 2016, 7, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Alarcon, B.; Mestre, D.; Martinez-Martin, N. The immunological synapse: A cause or consequence of T-cell receptor triggering? Immunology 2011, 133, 420–425. [Google Scholar] [CrossRef] [PubMed]
  134. Comrie, W.A.; Burkhardt, J.K. Action and Traction: Cytoskeletal Control of Receptor Triggering at the Immunological Synapse. Front. Immunol. 2016, 7, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Le Floc’h, A.; Huse, M. Molecular mechanisms and functional implications of polarized actin remodeling at the T cell immunological synapse. Cell. Mol. Life Sci. 2015, 72, 537–556. [Google Scholar] [CrossRef] [Green Version]
  136. Pettmann, J.; Santos, A.M.; Dushek, O.; Davis, S.J. Membrane Ultrastructure and T Cell Activation. Front. Immunol. 2018, 9, 2152. [Google Scholar] [CrossRef]
  137. Varga, G.; Nippe, N.; Balkow, S.; Peters, T.; Wild, M.K.; Seeliger, S.; Beissert, S.; Krummen, M.; Roth, J.; Sunderkotter, C.; et al. LFA-1 contributes to signal I of T-cell activation and to the production of T(h)1 cytokines. J. Investig. Dermatol. 2010, 130, 1005–1012. [Google Scholar] [CrossRef]
  138. Wang, Y.; Shibuya, K.; Yamashita, Y.; Shirakawa, J.; Shibata, K.; Kai, H.; Yokosuka, T.; Saito, T.; Honda, S.; Tahara-Hanaoka, S.; et al. LFA-1 decreases the antigen dose for T cell activation in vivo. Int. Immunol. 2008, 20, 1119–1127. [Google Scholar] [CrossRef]
  139. Palmer, E.; Drobek, A.; Stepanek, O. Opposing effects of actin signaling and LFA-1 on establishing the affinity threshold for inducing effector T-cell responses in mice. Eur. J. Immunol. 2016, 46, 1887–1901. [Google Scholar] [CrossRef] [Green Version]
  140. Hashimoto-Tane, A.; Sakuma, M.; Ike, H.; Yokosuka, T.; Kimura, Y.; Ohara, O.; Saito, T. Micro-adhesion rings surrounding TCR microclusters are essential for T cell activation. J. Exp. Med. 2016, 213, 1609–1625. [Google Scholar] [CrossRef]
  141. Azoulay-Alfaguter, I.; Strazza, M.; Peled, M.; Novak, H.K.; Muller, J.; Dustin, M.L.; Mor, A. The tyrosine phosphatase SHP-1 promotes T cell adhesion by activating the adaptor protein CrkII in the immunological synapse. Sci. Signal. 2017, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Kondo, N.; Ueda, Y.; Kita, T.; Ozawa, M.; Tomiyama, T.; Yasuda, K.; Lim, D.S.; Kinashi, T. NDR1-Dependent Regulation of Kindlin-3 Controls High-Affinity LFA-1 Binding and Immune Synapse Organization. Mol. Cell. Biol. 2017, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Garcon, F.; Okkenhaug, K. PI3Kdelta promotes CD4(+) T-cell interactions with antigen-presenting cells by increasing LFA-1 binding to ICAM-1. Immunol. Cell Biol. 2016, 94, 486–495. [Google Scholar] [CrossRef] [PubMed]
  144. Feigelson, S.W.; Grabovsky, V.; Manevich-Mendelson, E.; Pasvolsky, R.; Shulman, Z.; Shinder, V.; Klein, E.; Etzioni, A.; Aker, M.; Alon, R. Kindlin-3 is required for the stabilization of TCR-stimulated LFA-1:ICAM-1 bonds critical for lymphocyte arrest and spreading on dendritic cells. Blood 2011, 117, 7042–7052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Siokis, A.; Robert, P.A.; Demetriou, P.; Dustin, M.L.; Meyer-Hermann, M. F-Actin-Driven CD28-CD80 Localization in the Immune Synapse. Cell Rep. 2018, 24, 1151–1162. [Google Scholar] [CrossRef] [Green Version]
  146. Roy, N.H.; Burkhardt, J.K. The Actin Cytoskeleton: A Mechanical Intermediate for Signal Integration at the Immunological Synapse. Front. Cell Dev. Biol. 2018, 6, 116. [Google Scholar] [CrossRef] [Green Version]
  147. Murugesan, S.; Hong, J.; Yi, J.; Li, D.; Beach, J.R.; Shao, L.; Meinhardt, J.; Madison, G.; Wu, X.; Betzig, E.; et al. Formin-generated actomyosin arcs propel T cell receptor microcluster movement at the immune synapse. J. Cell Biol. 2016, 215, 383–399. [Google Scholar] [CrossRef]
  148. Santos, L.C.; Blair, D.A.; Kumari, S.; Cammer, M.; Iskratsch, T.; Herbin, O.; Alexandropoulos, K.; Dustin, M.L.; Sheetz, M.P. Actin polymerization-dependent activation of Cas-L promotes immunological synapse stability. Immunol. Cell Biol. 2016, 94, 981–993. [Google Scholar] [CrossRef] [Green Version]
  149. Borger, J.G.; Morrison, V.L.; Filby, A.; Garcia, C.; Uotila, L.M.; Simbari, F.; Fagerholm, S.C.; Zamoyska, R. Caveolin-1 Influences LFA-1 Redistribution upon TCR Stimulation in CD8 T Cells. J. Immunol. 2017, 199, 874–884. [Google Scholar] [CrossRef] [Green Version]
  150. Castro-Sanchez, P.; Ramirez-Munoz, R.; Martin-Cofreces, N.B.; Aguilar-Sopena, O.; Alegre-Gomez, S.; Hernandez-Perez, S.; Reyes, R.; Zeng, Q.; Cabanas, C.; Sanchez-Madrid, F.; et al. Phosphatase of Regenerating Liver-1 (PRL-1) Regulates Actin Dynamics During Immunological Synapse Assembly and T Cell Effector Function. Front. Immunol. 2018, 9, 2655. [Google Scholar] [CrossRef]
  151. Wabnitz, G.; Balta, E.; Samstag, Y. L-plastin regulates the stability of the immune synapse of naive and effector T-cells. Adv. Biol. Regul. 2017, 63, 107–114. [Google Scholar] [CrossRef] [PubMed]
  152. Na, B.R.; Jun, C.D. TAGLN2-mediated actin stabilization at the immunological synapse: Implication for cytotoxic T cell control of target cells. Bmb Rep. 2015, 48, 369–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Meissner, J.M.; Sikorski, A.F.; Nawara, T.; Grzesiak, J.; Marycz, K.; Boguslawska, D.M.; Michalczyk, I.; Lecomte, M.C.; Machnicka, B. alphaII-spectrin in T cells is involved in the regulation of cell-cell contact leading to immunological synapse formation? PLoS ONE 2017, 12, e0189545. [Google Scholar] [CrossRef] [Green Version]
  154. Jankowska, K.I.; Williamson, E.K.; Roy, N.H.; Blumenthal, D.; Chandra, V.; Baumgart, T.; Burkhardt, J.K. Integrins Modulate T Cell Receptor Signaling by Constraining Actin Flow at the Immunological Synapse. Front. Immunol. 2018, 9, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Markey, K.A.; Gartlan, K.H.; Kuns, R.D.; MacDonald, K.P.; Hill, G.R. Imaging the immunological synapse between dendritic cells and T cells. J. Immunol. Methods 2015, 423, 40–44. [Google Scholar] [CrossRef] [PubMed]
  156. Malinova, D.; Fritzsche, M.; Nowosad, C.R.; Armer, H.; Munro, P.M.; Blundell, M.P.; Charras, G.; Tolar, P.; Bouma, G.; Thrasher, A.J. WASp-dependent actin cytoskeleton stability at the dendritic cell immunological synapse is required for extensive, functional T cell contacts. J. Leukoc. Biol. 2016, 99, 699–710. [Google Scholar] [CrossRef] [PubMed]
  157. Wioland, H.; Guichard, B.; Senju, Y.; Myram, S.; Lappalainen, P.; Jegou, A.; Romet-Lemonne, G. ADF/Cofilin Accelerates Actin Dynamics by Severing Filaments and Promoting Their Depolymerization at Both Ends. Curr. Biol. Cb 2017, 27, 1956.e1957–1967.e1957. [Google Scholar] [CrossRef] [Green Version]
  158. Xu, Y.; Pektor, S.; Balkow, S.; Hemkemeyer, S.A.; Liu, Z.; Grobe, K.; Hanley, P.J.; Shen, L.; Bros, M.; Schmidt, T.; et al. Dendritic cell motility and T cell activation requires regulation of Rho-cofilin signaling by the Rho-GTPase activating protein myosin IXb. J. Immunol. 2014, 192, 3559–3568. [Google Scholar] [CrossRef] [Green Version]
  159. Comrie, W.A.; Li, S.; Boyle, S.; Burkhardt, J.K. The dendritic cell cytoskeleton promotes T cell adhesion and activation by constraining ICAM-1 mobility. J. Cell Biol. 2015, 208, 457–473. [Google Scholar] [CrossRef] [Green Version]
  160. Petit, A.E.; Demotte, N.; Scheid, B.; Wildmann, C.; Bigirimana, R.; Gordon-Alonso, M.; Carrasco, J.; Valitutti, S.; Godelaine, D.; van der Bruggen, P. A major secretory defect of tumour-infiltrating T lymphocytes due to galectin impairing LFA-1-mediated synapse completion. Nat. Commun. 2016, 7, 12242. [Google Scholar] [CrossRef] [Green Version]
  161. Ruvolo, P.P. Galectin 3 as a guardian of the tumor microenvironment. Biochim. Et Biophys. Acta 2016, 1863, 427–437. [Google Scholar] [CrossRef] [PubMed]
  162. Jeon, B.N.; Kim, H.R.; Chung, Y.S.; Na, B.R.; Park, H.; Hong, C.; Fatima, Y.; Oh, H.; Kim, C.H.; Jun, C.D. Actin stabilizer TAGLN2 potentiates adoptive T cell therapy by boosting the inside-out costimulation via lymphocyte function-associated antigen-1. Oncoimmunology 2018, 7, e1500674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Harris, E.S.; Weyrich, A.S.; Zimmerman, G.A. Lessons from rare maladies: Leukocyte adhesion deficiency syndromes. Curr. Opin. Hematol. 2013, 20, 16–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Levy-Mendelovich, S.; Rechavi, E.; Abuzaitoun, O.; Vernitsky, H.; Simon, A.J.; Lev, A.; Somech, R. Highlighting the problematic reliance on CD18 for diagnosing leukocyte adhesion deficiency type 1. Immunol Res. 2016, 64, 476–482. [Google Scholar] [CrossRef]
  165. Moutsopoulos, N.M.; Konkel, J.; Sarmadi, M.; Eskan, M.A.; Wild, T.; Dutzan, N.; Abusleme, L.; Zenobia, C.; Hosur, K.B.; Abe, T.; et al. Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven inflammatory bone loss. Sci. Transl. Med. 2014, 6, 229ra240. [Google Scholar] [CrossRef] [Green Version]
  166. Hajishengallis, G.; Moutsopoulos, N.M. Role of bacteria in leukocyte adhesion deficiency-associated periodontitis. Microb. Pathog. 2016, 94, 21–26. [Google Scholar] [CrossRef] [Green Version]
  167. Wolach, B.; Gavrieli, R.; Wolach, O.; Stauber, T.; Abuzaitoun, O.; Kuperman, A.; Amir, Y.; Stepensky, P.; Somech, R.; Etzioni, A. Leucocyte adhesion deficiency-A multicentre national experience. Eur. J. Clin. Investig. 2019, 49, e13047. [Google Scholar] [CrossRef]
  168. Almarza Novoa, E.; Kasbekar, S.; Thrasher, A.J.; Kohn, D.B.; Sevilla, J.; Nguyen, T.; Schwartz, J.D.; Bueren, J.A. Leukocyte adhesion deficiency-I: A comprehensive review of all published cases. J. Allergy Clin. Immunol Pr. 2018, 6, 1418–1420 e1410. [Google Scholar] [CrossRef]
  169. Giger, U.; Boxer, L.A.; Simpson, P.J.; Lucchesi, B.R.; Todd, R.F., 3rd. Deficiency of leukocyte surface glycoproteins Mo1, LFA-1, and Leu M5 in a dog with recurrent bacterial infections: An animal model. Blood 1987, 69, 1622–1630. [Google Scholar]
  170. Hanna, S.; Etzioni, A. Leukocyte adhesion deficiencies. Ann. New York Acad. Sci. 2012, 1250, 50–55. [Google Scholar] [CrossRef]
  171. Sturla, L.; Puglielli, L.; Tonetti, M.; Berninsone, P.; Hirschberg, C.B.; De Flora, A.; Etzioni, A. Impairment of the Golgi GDP-L-fucose transport and unresponsiveness to fucose replacement therapy in LAD II patients. Pediatr. Res. 2001, 49, 537–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Saultier, P.; Szepetowski, S.; Canault, M.; Falaise, C.; Poggi, M.; Suchon, P.; Barlogis, V.; Michel, G.; Loyau, S.; Jandrot-Perrus, M.; et al. Long-term management of leukocyte adhesion deficiency type III without hematopoietic stem cell transplantation. Haematologica 2018, 103, e264–e267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Suratannon, N.; Yeetong, P.; Srichomthong, C.; Amarinthnukrowh, P.; Chatchatee, P.; Sosothikul, D.; van Hagen, P.M.; van der Burg, M.; Wentink, M.; Driessen, G.J.; et al. Adaptive immune defects in a patient with leukocyte adhesion deficiency type III with a novel mutation in FERMT3. Pediatr. Allergy Immunol. 2016, 27, 214–217. [Google Scholar] [CrossRef] [PubMed]
  174. Stepensky, P.Y.; Wolach, B.; Gavrieli, R.; Rousso, S.; Ben Ami, T.; Goldman, V.; Rozovsky, K.; Hanna, S.; Etzioni, A.; Weintraub, M. Leukocyte adhesion deficiency type III: Clinical features and treatment with stem cell transplantation. J. Pediatr. Hematol. Oncol. 2015, 37, 264–268. [Google Scholar] [CrossRef] [PubMed]
  175. Wilson, R.W.; Ballantyne, C.M.; Smith, C.W.; Montgomery, C.; Bradley, A.; O’Brien, W.E.; Beaudet, A.L. Gene targeting yields a CD18-mutant mouse for study of inflammation. J. Immunol. 1993, 151, 1571–1578. [Google Scholar] [PubMed]
  176. Shimizu, K.; Libby, P.; Shubiki, R.; Sakuma, M.; Wang, Y.; Asano, K.; Mitchell, R.N.; Simon, D.I. Leukocyte integrin Mac-1 promotes acute cardiac allograft rejection. Circulation 2008, 117, 1997–2008. [Google Scholar] [CrossRef] [Green Version]
  177. Wang, H.; Peters, T.; Sindrilaru, A.; Scharffetter-Kochanek, K. Key role of macrophages in the pathogenesis of CD18 hypomorphic murine model of psoriasis. J. Investig. Dermatol. 2009, 129, 1100–1114. [Google Scholar] [CrossRef] [Green Version]
  178. Wang, H.; von Rohrscheidt, J.; Roehrbein, J.; Peters, T.; Sindrilaru, A.; Kess, D.; Preissner, K.T.; Scharffetter-Kochanek, K. Extracellular adherence protein of Staphylococcus aureus suppresses disease by inhibiting T-cell recruitment in a mouse model of psoriasis. J. Investig. Dermatol. 2010, 130, 743–754. [Google Scholar] [CrossRef] [Green Version]
  179. Scharffetter-Kochanek, K.; Lu, H.; Norman, K.; van Nood, N.; Munoz, F.; Grabbe, S.; McArthur, M.; Lorenzo, I.; Kaplan, S.; Ley, K.; et al. Spontaneous skin ulceration and defective T cell function in CD18 null mice. J. Exp. Med. 1998, 188, 119–131. [Google Scholar] [CrossRef] [Green Version]
  180. Leon-Rico, D.; Aldea, M.; Sanchez-Baltasar, R.; Mesa-Nunez, C.; Record, J.; Burns, S.O.; Santilli, G.; Thrasher, A.J.; Bueren, J.A.; Almarza, E. Lentiviral Vector-Mediated Correction of a Mouse Model of Leukocyte Adhesion Deficiency Type I. Hum. Gene 2016, 27, 668–678. [Google Scholar] [CrossRef] [Green Version]
  181. Sisco, M.; Chao, J.D.; Kim, I.; Mogford, J.E.; Mayadas, T.N.; Mustoe, T.A. Delayed wound healing in Mac-1-deficient mice is associated with normal monocyte recruitment. Wound Repair. Regen. 2007, 15, 566–571. [Google Scholar] [CrossRef] [PubMed]
  182. Meakin, P.J.; Morrison, V.L.; Sneddon, C.C.; Savinko, T.; Uotila, L.; Jalicy, S.M.; Gabriel, J.L.; Kang, L.; Ashford, M.L.; Fagerholm, S.C. Mice Lacking beta2-Integrin Function Remain Glucose Tolerant in Spite of Insulin Resistance, Neutrophil Infiltration and Inflammation. PLoS ONE 2015, 10, e0138872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Guilliams, M.; Ginhoux, F.; Jakubzick, C.; Naik, S.H.; Onai, N.; Schraml, B.U.; Segura, E.; Tussiwand, R.; Yona, S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat. Rev. Immunol. 2014, 14, 571–578. [Google Scholar] [CrossRef]
  184. Haasken, S.; Auger, J.L.; Binstadt, B.A. Absence of beta2 integrins impairs regulatory T cells and exacerbates CD4+ T cell-dependent autoimmune carditis. J. Immunol. 2011, 187, 2702–2710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Miura, Y.; Miura, M.; Gronthos, S.; Allen, M.R.; Cao, C.; Uveges, T.E.; Bi, Y.; Ehirchiou, D.; Kortesidis, A.; Shi, S.; et al. Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis. Proc. Natl. Acad. Sci. USA 2005, 102, 14022–14027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Bose, T.O.; Pham, Q.M.; Jellison, E.R.; Mouries, J.; Ballantyne, C.M.; Lefrancois, L. CD11a regulates effector CD8 T cell differentiation and central memory development in response to infection with Listeria monocytogenes. Infect. Immun. 2013, 81, 1140–1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Ghosh, S.; Chackerian, A.A.; Parker, C.M.; Ballantyne, C.M.; Behar, S.M. The LFA-1 adhesion molecule is required for protective immunity during pulmonary Mycobacterium tuberculosis infection. J. Immunol. 2006, 176, 4914–4922. [Google Scholar] [CrossRef] [Green Version]
  188. Gorina, R.; Lyck, R.; Vestweber, D.; Engelhardt, B. beta2 integrin-mediated crawling on endothelial ICAM-1 and ICAM-2 is a prerequisite for transcellular neutrophil diapedesis across the inflamed blood-brain barrier. J. Immunol. 2014, 192, 324–337. [Google Scholar] [CrossRef] [Green Version]
  189. Liu, X.; Jiang, X.; Liu, R.; Wang, L.; Qian, T.; Zheng, Y.; Deng, Y.; Huang, E.; Xu, F.; Wang, J.Y.; et al. B cells expressing CD11b effectively inhibit CD4+ T-cell responses and ameliorate experimental autoimmune hepatitis in mice. Hepatol. (Baltim. Md.) 2015, 62, 1563–1575. [Google Scholar] [CrossRef] [Green Version]
  190. Teschner, D.; Cholaszczynska, A.; Ries, F.; Beckert, H.; Theobald, M.; Grabbe, S.; Radsak, M.; Bros, M. CD11b Regulates Fungal Outgrowth but Not Neutrophil Recruitment in a Mouse Model of Invasive Pulmonary Aspergillosis. Front. Immunol. 2019, 10, 123. [Google Scholar] [CrossRef] [Green Version]
  191. Gao, X.P.; Liu, Q.; Broman, M.; Predescu, D.; Frey, R.S.; Malik, A.B. Inactivation of CD11b in a mouse transgenic model protects against sepsis-induced lung PMN infiltration and vascular injury. Physiol. Genom. 2005, 21, 230–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Coxon, A.; Rieu, P.; Barkalow, F.J.; Askari, S.; Sharpe, A.H.; von Andrian, U.H.; Arnaout, M.A.; Mayadas, T.N. A novel role for the beta 2 integrin CD11b/CD18 in neutrophil apoptosis: A homeostatic mechanism in inflammation. Immunity 1996, 5, 653–666. [Google Scholar] [CrossRef] [Green Version]
  193. Faridi, M.H.; Khan, S.Q.; Zhao, W.; Lee, H.W.; Altintas, M.M.; Zhang, K.; Kumar, V.; Armstrong, A.R.; Carmona-Rivera, C.; Dorschner, J.M.; et al. CD11b activation suppresses TLR-dependent inflammation and autoimmunity in systemic lupus erythematosus. J. Clin. Investig. 2017, 127, 1271–1283. [Google Scholar] [CrossRef] [Green Version]
  194. Wang, Y.; Gao, H.; Shi, C.; Erhardt, P.W.; Pavlovsky, A.; D, A.S.; Bledzka, K.; Ustinov, V.; Zhu, L.; Qin, J.; et al. Leukocyte integrin Mac-1 regulates thrombosis via interaction with platelet GPIbalpha. Nat. Commun. 2017, 8, 15559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Underhill, D.M.; Ozinsky, A. Phagocytosis of microbes: Complexity in action. Annu. Rev. Immunol. 2002, 20, 825–852. [Google Scholar] [CrossRef] [PubMed]
  196. Fu, H.; Liu, B.; Frost, J.L.; Hong, S.; Jin, M.; Ostaszewski, B.; Shankar, G.M.; Costantino, I.M.; Carroll, M.C.; Mayadas, T.N.; et al. Complement component C3 and complement receptor type 3 contribute to the phagocytosis and clearance of fibrillar Abeta by microglia. Glia 2012, 60, 993–1003. [Google Scholar] [CrossRef] [Green Version]
  197. Anderson, D.C.; Schmalsteig, F.C.; Finegold, M.J.; Hughes, B.J.; Rothlein, R.; Miller, L.J.; Kohl, S.; Tosi, M.F.; Jacobs, R.L.; Waldrop, T.C.; et al. The severe and moderate phenotypes of heritable Mac-1, LFA-1 deficiency: Their quantitative definition and relation to leukocyte dysfunction and clinical features. J. Infect. Dis. 1985, 152, 668–689. [Google Scholar] [CrossRef]
  198. Ley, K.; Hoffman, H.M.; Kubes, P.; Cassatella, M.A.; Zychlinsky, A.; Hedrick, C.C.; Catz, S.D. Neutrophils: New insights and open questions. Sci. Immunol. 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  199. BoseDasgupta, S.; Pieters, J. Macrophage-microbe interaction: Lessons learned from the pathogen Mycobacterium tuberculosis. Semin. Immunopathol. 2018, 40, 577–591. [Google Scholar] [CrossRef]
  200. Gazendam, R.P.; van de Geer, A.; Roos, D.; van den Berg, T.K.; Kuijpers, T.W. How neutrophils kill fungi. Immunol. Rev. 2016, 273, 299–311. [Google Scholar] [CrossRef]
  201. Lu, H.; Ballantyne, C.; Smith, C.W. LFA-1 (CD11a/CD18) triggers hydrogen peroxide production by canine neutrophils. J. Leukoc. Biol. 2000, 68, 73–80. [Google Scholar] [PubMed]
  202. Allen, S.J.; Mott, K.R.; Chentoufi, A.A.; BenMohamed, L.; Wechsler, S.L.; Ballantyne, C.M.; Ghiasi, H. CD11c controls herpes simplex virus 1 responses to limit virus replication during primary infection. J. Virol. 2011, 85, 9945–9955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Spear, P.G. Herpes simplex virus: Receptors and ligands for cell entry. Cell. Microbiol. 2004, 6, 401–410. [Google Scholar] [CrossRef] [PubMed]
  204. Emoto, M.; Emoto, Y.; Brinkmann, V.; Miyamoto, M.; Yoshizawa, I.; Staber, M.; van Rooijen, N.; Hamann, A.; Kaufmann, S.H. Increased resistance of LFA-1-deficient mice to lipopolysaccharide-induced shock/liver injury in the presence of TNF-alpha and IL-12 is mediated by IL-10: A novel role for LFA-1 in the regulation of the proinflammatory and anti-inflammatory cytokine balance. J. Immunol. 2003, 171, 584–593. [Google Scholar] [CrossRef] [PubMed]
  205. Basit, A.; Reutershan, J.; Morris, M.A.; Solga, M.; Rose, C.E., Jr.; Ley, K. ICAM-1 and LFA-1 play critical roles in LPS-induced neutrophil recruitment into the alveolar space. Am. J. Physiol. Lung Cell. Mol. Physiol. 2006, 291, L200–L207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Liu, J.R.; Han, X.; Soriano, S.G.; Yuki, K. The role of macrophage 1 antigen in polymicrobial sepsis. Shock (Augustaga.) 2014, 42, 532–539. [Google Scholar] [CrossRef]
  207. Echtenacher, B.; Mannel, D.N.; Hultner, L. Critical protective role of mast cells in a model of acute septic peritonitis. Nature 1996, 381, 75–77. [Google Scholar] [CrossRef]
  208. Rosenkranz, A.R.; Coxon, A.; Maurer, M.; Gurish, M.F.; Austen, K.F.; Friend, D.S.; Galli, S.J.; Mayadas, T.N. Impaired mast cell development and innate immunity in Mac-1 (CD11b/CD18, CR3)-deficient mice. J. Immunol. 1998, 161, 6463–6467. [Google Scholar]
  209. Hawkins, H.K.; Heffelfinger, S.C.; Anderson, D.C. Leukocyte adhesion deficiency: Clinical and postmortem observations. Pediatric Pathol. 1992, 12, 119–130. [Google Scholar] [CrossRef]
  210. Mizgerd, J.P.; Horwitz, B.H.; Quillen, H.C.; Scott, M.L.; Doerschuk, C.M. Effects of CD18 deficiency on the emigration of murine neutrophils during pneumonia. J. Immunol. 1999, 163, 995–999. [Google Scholar]
  211. Rijneveld, A.W.; de Vos, A.F.; Florquin, S.; Verbeek, J.S.; van der Poll, T. CD11b limits bacterial outgrowth and dissemination during murine pneumococcal pneumonia. J. Infect. Dis. 2005, 191, 1755–1760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Hu, C.; Mayadas-Norton, T.; Tanaka, K.; Chan, J.; Salgame, P. Mycobacterium tuberculosis infection in complement receptor 3-deficient mice. J. Immunol. 2000, 165, 2596–2602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Mysak, J.; Podzimek, S.; Sommerova, P.; Lyuya-Mi, Y.; Bartova, J.; Janatova, T.; Prochazkova, J.; Duskova, J. Porphyromonas gingivalis: Major periodontopathic pathogen overview. J. Immunol. Res. 2014, 2014, 476068. [Google Scholar] [CrossRef] [Green Version]
  214. Wang, M.; Shakhatreh, M.A.; James, D.; Liang, S.; Nishiyama, S.; Yoshimura, F.; Demuth, D.R.; Hajishengallis, G. Fimbrial proteins of porphyromonas gingivalis mediate in vivo virulence and exploit TLR2 and complement receptor 3 to persist in macrophages. J. Immunol. 2007, 179, 2349–2358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Zundler, S.; Neurath, M.F. Interleukin-12: Functional activities and implications for disease. Cytokine Growth Factor Rev. 2015, 26, 559–568. [Google Scholar] [CrossRef] [PubMed]
  216. Hajishengallis, G.; Shakhatreh, M.A.; Wang, M.; Liang, S. Complement receptor 3 blockade promotes IL-12-mediated clearance of Porphyromonas gingivalis and negates its virulence in vivo. J. Immunol. 2007, 179, 2359–2367. [Google Scholar] [CrossRef] [Green Version]
  217. Radoshevich, L.; Cossart, P. Listeria monocytogenes: Towards a complete picture of its physiology and pathogenesis. Nat. Rev. Microbiol. 2018, 16, 32–46. [Google Scholar] [CrossRef]
  218. Wu, H.; Prince, J.E.; Brayton, C.F.; Shah, C.; Zeve, D.; Gregory, S.H.; Smith, C.W.; Ballantyne, C.M. Host resistance of CD18 knockout mice against systemic infection with Listeria monocytogenes. Infect. Immun. 2003, 71, 5986–5993. [Google Scholar] [CrossRef] [Green Version]
  219. Miyamoto, M.; Emoto, M.; Emoto, Y.; Brinkmann, V.; Yoshizawa, I.; Seiler, P.; Aichele, P.; Kita, E.; Kaufmann, S.H. Neutrophilia in LFA-1-deficient mice confers resistance to listeriosis: Possible contribution of granulocyte-colony-stimulating factor and IL-17. J. Immunol. 2003, 170, 5228–5234. [Google Scholar] [CrossRef]
  220. Roberts, A.W. G-CSF: A key regulator of neutrophil production, but that’s not all! Growth Factors (Churswitzerland) 2005, 23, 33–41. [Google Scholar] [CrossRef]
  221. Emoto, M.; Miyamoto, M.; Emoto, Y.; Yoshizawa, I.; Brinkmann, V.; van Rooijen, N.; Kaufmann, S.H. Highly biased type 1 immune responses in mice deficient in LFA-1 in Listeria monocytogenes infection are caused by elevated IL-12 production by granulocytes. J. Immunol. 2003, 171, 3970–3976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Gregory, S.H.; Cousens, L.P.; van Rooijen, N.; Dopp, E.A.; Carlos, T.M.; Wing, E.J. Complementary adhesion molecules promote neutrophil-Kupffer cell interaction and the elimination of bacteria taken up by the liver. J. Immunol. 2002, 168, 308–315. [Google Scholar] [CrossRef] [PubMed]
  223. Guerau-de-Arellano, M.; Alroy, J.; Bullard, D.; Huber, B.T. Aggravated Lyme carditis in CD11a-/- and CD11c-/- mice. Infect. Immun. 2005, 73, 7637–7643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Nascimento, D.O.; Vieira-de-Abreu, A.; Arcanjo, A.F.; Bozza, P.T.; Zimmerman, G.A.; Castro-Faria-Neto, H.C. Integrin alphaDbeta2 (CD11d/CD18) Modulates Leukocyte Accumulation, Pathogen Clearance, and Pyroptosis in Experimental Salmonella Typhimurium Infection. Front. Immunol. 2018, 9, 1128. [Google Scholar] [CrossRef] [PubMed]
  225. Gong, W.; Shi, Y.; Ren, J. Research progresses of molecular mechanism of pyroptosis and its related diseases. Immunobiology 2019. [Google Scholar] [CrossRef]
  226. Gazendam, R.P.; van Hamme, J.L.; Tool, A.T.; Hoogenboezem, M.; van den Berg, J.M.; Prins, J.M.; Vitkov, L.; van de Veerdonk, F.L.; van den Berg, T.K.; Roos, D.; et al. Human Neutrophils Use Different Mechanisms to Kill Aspergillus fumigatus Conidia and Hyphae: Evidence from Phagocyte Defects. J. Immunol. 2016, 196, 1272–1283. [Google Scholar] [CrossRef] [Green Version]
  227. Rohm, M.; Grimm, M.J.; D’Auria, A.C.; Almyroudis, N.G.; Segal, B.H.; Urban, C.F. NADPH oxidase promotes neutrophil extracellular trap formation in pulmonary aspergillosis. Infect. Immun. 2014, 82, 1766–1777. [Google Scholar] [CrossRef] [Green Version]
  228. Urban, C.F.; Nett, J.E. Neutrophil extracellular traps in fungal infection. Semin. Cell Dev. Biol. 2019, 89, 47–57. [Google Scholar] [CrossRef]
  229. Clark, H.L.; Abbondante, S.; Minns, M.S.; Greenberg, E.N.; Sun, Y.; Pearlman, E. Protein Deiminase 4 and CR3 Regulate Aspergillus fumigatus and beta-Glucan-Induced Neutrophil Extracellular Trap Formation, but Hyphal Killing Is Dependent Only on CR3. Front. Immunol. 2018, 9, 1182. [Google Scholar] [CrossRef]
  230. Schmiedel, Y.; Zimmerli, S. Common invasive fungal diseases: An overview of invasive candidiasis, aspergillosis, cryptococcosis, and Pneumocystis pneumonia. Swiss Med. Wkly. 2016, 146, w14281. [Google Scholar] [CrossRef] [Green Version]
  231. O’Brien, X.M.; Reichner, J.S. Neutrophil Integrins and Matrix Ligands and NET Release. Front. Immunol. 2016, 7, 363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Gazendam, R.P.; van Hamme, J.L.; Tool, A.T.; van Houdt, M.; Verkuijlen, P.J.; Herbst, M.; Liese, J.G.; van de Veerdonk, F.L.; Roos, D.; van den Berg, T.K.; et al. Two independent killing mechanisms of Candida albicans by human neutrophils: Evidence from innate immunity defects. Blood 2014, 124, 590–597. [Google Scholar] [CrossRef] [PubMed]
  233. Soloviev, D.A.; Jawhara, S.; Fonzi, W.A. Regulation of innate immune response to Candida albicans infections by alphaMbeta2-Pra1p interaction. Infect. Immun. 2011, 79, 1546–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Ghorbani, M.; Farhoudi, R. Leishmaniasis in humans: Drug or vaccine therapy? Drug Des. Dev. Ther. 2018, 12, 25–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Schonlau, F.; Scharffetter-Kochanek, K.; Grabbe, S.; Pietz, B.; Sorg, C.; Sunderkotter, C. In experimental leishmaniasis deficiency of CD18 results in parasite dissemination associated with altered macrophage functions and incomplete Th1 cell response. Eur. J. Immunol. 2000, 30, 2729–2740. [Google Scholar] [CrossRef]
  236. PrabhuDas, M.R.; Baldwin, C.L.; Bollyky, P.L.; Bowdish, D.M.E.; Drickamer, K.; Febbraio, M.; Herz, J.; Kobzik, L.; Krieger, M.; Loike, J.; et al. A Consensus Definitive Classification of Scavenger Receptors and Their Roles in Health and Disease. J. Immunol. 2017, 198, 3775–3789. [Google Scholar] [CrossRef] [Green Version]
  237. Olekhnovitch, R.; Bousso, P. Induction, Propagation, and Activity of Host Nitric Oxide: Lessons from Leishmania Infection. Trends Parasitol. 2015, 31, 653–664. [Google Scholar] [CrossRef]
  238. Ricardo-Carter, C.; Favila, M.; Polando, R.E.; Cotton, R.N.; Bogard Horner, K.; Condon, D.; Ballhorn, W.; Whitcomb, J.P.; Yadav, M.; Geister, R.L.; et al. Leishmania major inhibits IL-12 in macrophages by signalling through CR3 (CD11b/CD18) and down-regulation of ETS-mediated transcription. Parasite Immunol. 2013, 35, 409–420. [Google Scholar] [CrossRef] [Green Version]
  239. Carter, C.R.; Whitcomb, J.P.; Campbell, J.A.; Mukbel, R.M.; McDowell, M.A. Complement receptor 3 deficiency influences lesion progression during Leishmania major infection in BALB/c mice. Infect. Immun. 2009, 77, 5668–5675. [Google Scholar] [CrossRef] [Green Version]
  240. Okwor, I.; Uzonna, J.E. Pathways leading to interleukin-12 production and protective immunity in cutaneous leishmaniasis. Cell. Immunol. 2016, 309, 32–36. [Google Scholar] [CrossRef]
  241. Miyazaki, Y.; Bunting, M.; Stafforini, D.M.; Harris, E.S.; McIntyre, T.M.; Prescott, S.M.; Frutuoso, V.S.; Amendoeira, F.C.; de Oliveira Nascimento, D.; Vieira-de-Abreu, A.; et al. Integrin alphaDbeta2 is dynamically expressed by inflamed macrophages and alters the natural history of lethal systemic infections. J. Immunol. 2008, 180, 590–600. [Google Scholar] [CrossRef] [Green Version]
  242. Perez-Mazliah, D.; Langhorne, J. CD4 T-cell subsets in malaria: TH1/TH2 revisited. Front. Immunol. 2014, 5, 671. [Google Scholar] [CrossRef] [PubMed]
  243. de Azevedo-Quintanilha, I.G.; Vieira-de-Abreu, A.; Ferreira, A.C.; Nascimento, D.O.; Siqueira, A.M.; Campbell, R.A.; Teixeira Ferreira, T.P.; Gutierrez, T.M.; Ribeiro, G.M. Integrin alphaDbeta2 (CD11d/CD18) mediates experimental malaria-associated acute respiratory distress syndrome (MA-ARDS). Malar. J. 2016, 15, 393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Franz, J.; Tarantola, M.; Riethmuller, C. How tetraspanins shape endothelial and leukocyte nano-architecture during inflammation. Biochem. Soc. Trans. 2017, 45, 999–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Patten, D.A.; Shetty, S. More Than Just a Removal Service: Scavenger Receptors in Leukocyte Trafficking. Front. Immunol. 2018, 9, 2904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Ivetic, A.; Hoskins Green, H.L.; Hart, S.J. L-selectin: A Major Regulator of Leukocyte Adhesion, Migration and Signaling. Front. Immunol. 2019, 10, 1068. [Google Scholar] [CrossRef] [Green Version]
  247. Klann, J.E.; Kim, S.H.; Remedios, K.A.; He, Z.; Metz, P.J.; Lopez, J.; Tysl, T.; Olvera, J.G.; Ablack, J.N.; Cantor, J.M.; et al. Integrin Activation Controls Regulatory T Cell-Mediated Peripheral Tolerance. J. Immunol. 2018, 200, 4012–4023. [Google Scholar] [CrossRef] [Green Version]
  248. Lewis, K.L.; Reizis, B. Dendritic cells: Arbiters of immunity and immunological tolerance. Cold Spring Harb. Perspect. Biol. 2012, 4, a007401. [Google Scholar] [CrossRef] [Green Version]
  249. Ehirchiou, D.; Xiong, Y.; Xu, G.; Chen, W.; Shi, Y.; Zhang, L. CD11b facilitates the development of peripheral tolerance by suppressing Th17 differentiation. J. Exp. Med. 2007, 204, 1519–1524. [Google Scholar] [CrossRef] [Green Version]
  250. Arnaout, M.A. Biology and structure of leukocyte beta 2 integrins and their role in inflammation. F1000Res 2016, 5. [Google Scholar] [CrossRef]
  251. Subramanian, P.; Mitroulis, I.; Hajishengallis, G.; Chavakis, T. Regulation of tissue infiltration by neutrophils: Role of integrin alpha3beta1 and other factors. Curr. Opin. Hematol. 2016, 23, 36–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. De Rose, D.U.; Giliani, S.; Notarangelo, L.D.; Lougaris, V.; Lanfranchi, A.; Moratto, D.; Martire, B.; Specchia, F.; Tommasini, A.; Plebani, A.; et al. Long term outcome of eight patients with type 1 Leukocyte Adhesion Deficiency (LAD-1): Not only infections, but high risk of autoimmune complications. Clin. Immunol. 2018, 191, 75–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. D’Agata, I.D.; Paradis, K.; Chad, Z.; Bonny, Y.; Seidman, E. Leucocyte adhesion deficiency presenting as a chronic ileocolitis. Gut 1996, 39, 605–608. [Google Scholar] [CrossRef] [PubMed]
  254. Uzel, G.; Kleiner, D.E.; Kuhns, D.B.; Holland, S.M. Dysfunctional LAD-1 neutrophils and colitis. Gastroenterology 2001, 121, 958–964. [Google Scholar] [CrossRef]
  255. Stevanin, M.; Busso, N.; Chobaz, V.; Pigni, M.; Ghassem-Zadeh, S.; Zhang, L.; Acha-Orbea, H.; Ehirchiou, D. CD11b regulates the Treg/Th17 balance in murine arthritis via IL-6. Eur. J. Immunol. 2017, 47, 637–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Van Seventer, G.A.; Shimizu, Y.; Horgan, K.J.; Shaw, S. The LFA-1 ligand ICAM-1 provides an important costimulatory signal for T cell receptor-mediated activation of resting T cells. J. Immunol. 1990, 144, 4579–4586. [Google Scholar]
  257. Constantinescu, C.S.; Farooqi, N.; O’Brien, K.; Gran, B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br. J. Pharmacol. 2011, 164, 1079–1106. [Google Scholar] [CrossRef] [PubMed]
  258. Gultner, S.; Kuhlmann, T.; Hesse, A.; Weber, J.P.; Riemer, C.; Baier, M.; Hutloff, A. Reduced Treg frequency in LFA-1-deficient mice allows enhanced T effector differentiation and pathology in EAE. Eur. J. Immunol. 2010, 40, 3403–3412. [Google Scholar] [CrossRef] [Green Version]
  259. Fu, J.H.; Zhou, C.C.; Mu, H.Q.; Nan, C.J.; Li, S.; Lu, D.Q. CD18 inhibits progression of kidney cancer by down-regulating Treg cell levels. Eur. Rev. Med. Pharm. Sci. 2019, 23, 2750–2755. [Google Scholar] [CrossRef]
  260. Koboziev, I.; Karlsson, F.; Ostanin, D.V.; Gray, L.; Davidson, M.; Zhang, S.; Grisham, M.B. Role of LFA-1 in the activation and trafficking of T cells: Implications in the induction of chronic colitis. Inflamm. Bowel Dis. 2012, 18, 2360–2370. [Google Scholar] [CrossRef] [Green Version]
  261. Dugger, K.J.; Zinn, K.R.; Weaver, C.; Bullard, D.C.; Barnum, S.R. Effector and suppressor roles for LFA-1 during the development of experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2009, 206, 22–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Bravo, B.; Gallego, M.I.; Flores, A.I.; Bornstein, R.; Puente-Bedia, A.; Hernandez, J.; de la Torre, P.; Garcia-Zaragoza, E.; Perez-Tavarez, R.; Grande, J.; et al. Restrained Th17 response and myeloid cell infiltration into the central nervous system by human decidua-derived mesenchymal stem cells during experimental autoimmune encephalomyelitis. Stem Cell Res. 2016, 7, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Gensterblum, E.; Renauer, P.; Coit, P.; Strickland, F.M.; Kilian, N.C.; Miller, S.; Ognenovski, M.; Wren, J.D.; Tsou, P.S.; Lewis, E.E.; et al. CD4+CD28+KIR+CD11a(hi) T cells correlate with disease activity and are characterized by a pro-inflammatory epigenetic and transcriptional profile in lupus patients. J. Autoimmun. 2018, 86, 19–28. [Google Scholar] [CrossRef]
  264. Wang, Y.; Shu, Y.; Xiao, Y.; Wang, Q.; Kanekura, T.; Li, Y.; Wang, J.; Zhao, M.; Lu, Q.; Xiao, R. Hypomethylation and overexpression of ITGAL (CD11a) in CD4(+) T cells in systemic sclerosis. Clin. Epigenetics 2014, 6, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Liu, Y.X.; Zhang, F.; Yao, Q.M.; Yuan, T.; Xu, J.; Zhu, X.J. Expression of CD11a in lymphocyte subpopulation in immune thrombocytopenia. Int. J. Clin. Exp. Pathol. 2015, 8, 15642–15651. [Google Scholar] [PubMed]
  266. Guttman-Yassky, E.; Vugmeyster, Y.; Lowes, M.A.; Chamian, F.; Kikuchi, T.; Kagen, M.; Gilleaudeau, P.; Lee, E.; Hunte, B.; Howell, K.; et al. Blockade of CD11a by efalizumab in psoriasis patients induces a unique state of T-cell hyporesponsiveness. J. Investig. Dermatol. 2008, 128, 1182–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Schwab, N.; Ulzheimer, J.C.; Fox, R.J.; Schneider-Hohendorf, T.; Kieseier, B.C.; Monoranu, C.M.; Staugaitis, S.M.; Welch, W.; Jilek, S.; Du Pasquier, R.A.; et al. Fatal PML associated with efalizumab therapy: Insights into integrin alphaLbeta2 in JC virus control. Neurology 2012, 78, 458–467. [Google Scholar] [CrossRef] [Green Version]
  268. Sterry, W.; Bagot, M.; Ferrandiz, C.; Kragballe, K.; Papp, K.; Stingl, G. Immunosuppressive therapy in dermatology and PML. J. Dtsch. Derm. Ges. 2009, 7, 5. [Google Scholar] [CrossRef] [Green Version]
  269. Bullard, D.C.; Hu, X.; Schoeb, T.R.; Axtell, R.C.; Raman, C.; Barnum, S.R. Critical requirement of CD11b (Mac-1) on T cells and accessory cells for development of experimental autoimmune encephalomyelitis. J. Immunol. 2005, 175, 6327–6333. [Google Scholar] [CrossRef] [Green Version]
  270. Rosetti, F.; Tsuboi, N.; Chen, K.; Nishi, H.; Ernandez, T.; Sethi, S.; Croce, K.; Stavrakis, G.; Alcocer-Varela, J.; Gomez-Martin, D.; et al. Human lupus serum induces neutrophil-mediated organ damage in mice that is enabled by Mac-1 deficiency. J. Immunol. 2012, 189, 3714–3723. [Google Scholar] [CrossRef] [Green Version]
  271. McAdoo, S.P.; Pusey, C.D. Antiglomerular Basement Membrane Disease. Semin. Respir. Crit. Care Med. 2018, 39, 494–503. [Google Scholar] [CrossRef] [PubMed]
  272. Tang, T.; Rosenkranz, A.; Assmann, K.J.; Goodman, M.J.; Gutierrez-Ramos, J.C.; Carroll, M.C.; Cotran, R.S.; Mayadas, T.N. A role for Mac-1 (CDIIb/CD18) in immune complex-stimulated neutrophil function in vivo: Mac-1 deficiency abrogates sustained Fcgamma receptor-dependent neutrophil adhesion and complement-dependent proteinuria in acute glomerulonephritis. J. Exp. Med. 1997, 186, 1853–1863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Bernard, P.; Antonicelli, F. Bullous Pemphigoid: A Review of its Diagnosis, Associations and Treatment. Am. J. Clin. Dermatol. 2017, 18, 513–528. [Google Scholar] [CrossRef] [PubMed]
  274. Zhang, Y.; Hwang, B.J.; Liu, Z.; Li, N.; Lough, K.; Williams, S.E.; Chen, J.; Burette, S.W.; Diaz, L.A.; Su, M.A.; et al. BP180 dysfunction triggers spontaneous skin inflammation in mice. Proc. Natl. Acad. Sci. USA 2018, 115, 6434–6439. [Google Scholar] [CrossRef] [Green Version]
  275. Liu, Z.; Zhao, M.; Li, N.; Diaz, L.A.; Mayadas, T.N. Differential roles for beta2 integrins in experimental autoimmune bullous pemphigoid. Blood 2006, 107, 1063–1069. [Google Scholar] [CrossRef] [Green Version]
  276. Lee, Y.H.; Bae, S.C. Association between the functional ITGAM rs1143679 G/A polymorphism and systemic lupus erythematosus/lupus nephritis or rheumatoid arthritis: An update meta-analysis. Rheumatol. Int. 2015, 35, 815–823. [Google Scholar] [CrossRef]
  277. Toller-Kawahisa, J.E.; Vigato-Ferreira, I.C.; Pancoto, J.A.; Mendes-Junior, C.T.; Martinez, E.Z.; Palomino, G.M.; Louzada-Junior, P.; Donadi, E.A.; Del Lama, J.E.; Marzocchi-Machado, C.M. The variant of CD11b, rs1143679 within ITGAM, is associated with systemic lupus erythematosus and clinical manifestations in Brazilian patients. Hum. Immunol. 2014, 75, 119–123. [Google Scholar] [CrossRef]
  278. Li, C.; Tong, F.; Ma, Y.; Qian, K.; Zhang, J.; Chen, X. Association of the CD11b rs1143679 polymorphism with systemic lupus erythematosus in the Han Chinese population. J. Int. Med. Res. 2018, 46, 1008–1014. [Google Scholar] [CrossRef]
  279. Nieuwenhuizen, N.E.; Kirstein, F.; Hoving, J.C.; Brombacher, F. House dust mite induced allergic airway disease is attenuated in CD11c(cre)IL-4Ralpha(-/l) degrees (x) mice. Sci. Rep. 2018, 8, 885. [Google Scholar] [CrossRef] [Green Version]
  280. Sadhu, C.; Ting, H.J.; Lipsky, B.; Hensley, K.; Garcia-Martinez, L.F.; Simon, S.I.; Staunton, D.E. CD11c/CD18: Novel ligands and a role in delayed-type hypersensitivity. J. Leukoc. Biol. 2007, 81, 1395–1403. [Google Scholar] [CrossRef]
  281. Miyashita, M.; Sasano, H.; Tamaki, K.; Hirakawa, H.; Takahashi, Y.; Nakagawa, S.; Watanabe, G.; Tada, H.; Suzuki, A.; Ohuchi, N.; et al. Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: A retrospective multicenter study. Breast Cancer Res. 2015, 17, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Barbier, L.; Ferhat, M.; Salame, E.; Robin, A.; Herbelin, A.; Gombert, J.M.; Silvain, C.; Barbarin, A. Interleukin-1 Family Cytokines: Keystones in Liver Inflammatory Diseases. Front. Immunol. 2019, 10, 2014. [Google Scholar] [CrossRef] [PubMed]
  283. Glatigny, S.; Duhen, R.; Arbelaez, C.; Kumari, S.; Bettelli, E. Integrin alpha L controls the homing of regulatory T cells during CNS autoimmunity in the absence of integrin alpha 4. Sci. Rep. 2015, 5, 7834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Luo, Y.; Wang, Y.; Shu, Y.; Lu, Q.; Xiao, R. Epigenetic mechanisms: An emerging role in pathogenesis and its therapeutic potential in systemic sclerosis. Int. J. Biochem. Cell Biol. 2015, 67, 92–100. [Google Scholar] [CrossRef]
  285. Sanchez-Blanco, C.; Clarke, F.; Cornish, G.H.; Depoil, D.; Thompson, S.J.; Dai, X.; Rawlings, D.J.; Dustin, M.L.; Zamoyska, R.; Cope, A.P.; et al. Protein tyrosine phosphatase PTPN22 regulates LFA-1 dependent Th1 responses. J. Autoimmun. 2018, 94, 45–55. [Google Scholar] [CrossRef]
  286. Singh, K.; Colmegna, I.; He, X.; Weyand, C.M.; Goronzy, J.J. Synoviocyte stimulation by the LFA-1-intercellular adhesion molecule-2-Ezrin-Akt pathway in rheumatoid arthritis. J. Immunol. 2008, 180, 1971–1978. [Google Scholar] [CrossRef] [Green Version]
  287. Watts, G.M.; Beurskens, F.J.; Martin-Padura, I.; Ballantyne, C.M.; Klickstein, L.B.; Brenner, M.B.; Lee, D.M. Manifestations of inflammatory arthritis are critically dependent on LFA-1. J. Immunol. 2005, 174, 3668–3675. [Google Scholar] [CrossRef]
  288. Esterhazy, D.; Loschko, J.; London, M.; Jove, V.; Oliveira, T.Y.; Mucida, D. Classical dendritic cells are required for dietary antigen-mediated induction of peripheral T(reg) cells and tolerance. Nat. Immunol. 2016, 17, 545–555. [Google Scholar] [CrossRef]
  289. Fagerholm, S.C.; MacPherson, M.; James, M.J.; Sevier-Guy, C.; Lau, C.S. The CD11b-integrin (ITGAM) and systemic lupus erythematosus. Lupus 2013, 22, 657–663. [Google Scholar] [CrossRef]
  290. Rosetti, F.; de la Cruz, A.; Crispin, J.C. Gene-function studies in systemic lupus erythematosus. Curr. Opin. Rheumatol. 2019, 31, 185–192. [Google Scholar] [CrossRef]
  291. Wang, L.; Li, Z.; Ciric, B.; Safavi, F.; Zhang, G.X.; Rostami, A. Selective depletion of CD11c(+) CD11b(+) dendritic cells partially abrogates tolerogenic effects of intravenous MOG in murine EAE. Eur. J. Immunol. 2016, 46, 2454–2466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Besusso, D.; Saul, L.; Leech, M.D.; O’Connor, R.A.; MacDonald, A.S.; Anderton, S.M.; Mellanby, R.J. 1,25-Dihydroxyvitamin D3-Conditioned CD11c+ Dendritic Cells are Effective Initiators of CNS Autoimmune Disease. Front. Immunol. 2015, 6, 575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Cao, L.Y.; Soler, D.C.; Debanne, S.M.; Grozdev, I.; Rodriguez, M.E.; Feig, R.L.; Carman, T.L.; Gilkeson, R.C.; Orringer, C.E.; Kern, E.F.; et al. Psoriasis and cardiovascular risk factors: Increased serum myeloperoxidase and corresponding immunocellular overexpression by Cd11b(+) CD68(+) macrophages in skin lesions. Am. J. Transl. Res. 2013, 6, 16–27. [Google Scholar] [PubMed]
  294. van Pelt, J.P.; Kuijpers, S.H.; van de Kerkhof, P.C.; de Jong, E.M. The CD11b/CD18-integrin in the pathogenesis of psoriasis. J. Derm. Sci. 1998, 16, 135–143. [Google Scholar] [CrossRef]
  295. Sjogren, F.; Ljunghusen, O.; Baas, A.; Coble, B.I.; Stendahl, O. Expression and function of beta 2 integrin CD11B/CD18 on leukocytes from patients with psoriasis. Acta Derm. Venereol. 1999, 79, 105–110. [Google Scholar] [CrossRef]
  296. Ito, A.; Hong, C.; Oka, K.; Salazar, J.V.; Diehl, C.; Witztum, J.L.; Diaz, M.; Castrillo, A.; Bensinger, S.J.; Chan, L.; et al. Cholesterol Accumulation in CD11c(+) Immune Cells Is a Causal and Targetable Factor in Autoimmune Disease. Immunity 2016, 45, 1311–1326. [Google Scholar] [CrossRef] [Green Version]
  297. Thomas, A.P.; Dunn, T.N.; Oort, P.J.; Grino, M.; Adams, S.H. Inflammatory phenotyping identifies CD11d as a gene markedly induced in white adipose tissue in obese rodents and women. J. Nutr. 2011, 141, 1172–1180. [Google Scholar] [CrossRef] [Green Version]
  298. Lanitis, E.; Dangaj, D.; Irving, M.; Coukos, G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann. Oncol. 2017, 28, xii18–xii32. [Google Scholar] [CrossRef]
  299. Prenen, H.; Mazzone, M. Tumor-associated macrophages: A short compendium. Cell. Mol. Life Sci. 2019, 76, 1447–1458. [Google Scholar] [CrossRef]
  300. Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef]
  301. Schupp, J.; Krebs, F.K.; Zimmer, N.; Trzeciak, E.; Schuppan, D.; Tuettenberg, A. Targeting myeloid cells in the tumor sustaining microenvironment. Cell. Immunol. 2019, 343, 103713. [Google Scholar] [CrossRef] [PubMed]
  302. Tcyganov, E.; Mastio, J.; Chen, E.; Gabrilovich, D.I. Plasticity of myeloid-derived suppressor cells in cancer. Curr. Opin. Immunol. 2018, 51, 76–82. [Google Scholar] [CrossRef] [PubMed]
  303. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Harjunpaa, H.; Llort Asens, M.; Guenther, C.; Fagerholm, S.C. Cell Adhesion Molecules and Their Roles and Regulation in the Immune and Tumor Microenvironment. Front. Immunol. 2019, 10, 1078. [Google Scholar] [CrossRef] [Green Version]
  305. Zhang, Q.Q.; Hu, X.W.; Liu, Y.L.; Ye, Z.J.; Gui, Y.H.; Zhou, D.L.; Qi, C.L.; He, X.D.; Wang, H.; Wang, L.J. CD11b deficiency suppresses intestinal tumor growth by reducing myeloid cell recruitment. Sci. Rep. 2015, 5, 15948. [Google Scholar] [CrossRef]
  306. Deronic, A.; Tahvili, S.; Leanderson, T.; Ivars, F. The anti-tumor effect of the quinoline-3-carboxamide tasquinimod: Blockade of recruitment of CD11b(+) Ly6C(hi) cells to tumor tissue reduces tumor growth. Bmc Cancer 2016, 16, 440. [Google Scholar] [CrossRef] [Green Version]
  307. Okita, Y.; Tanaka, H.; Ohira, M.; Muguruma, K.; Kubo, N.; Watanabe, M.; Fukushima, W.; Hirakawa, K. Role of tumor-infiltrating CD11b+ antigen-presenting cells in the progression of gastric cancer. J. Surg. Res. 2014, 186, 192–200. [Google Scholar] [CrossRef] [Green Version]
  308. Kim, K.J.; Lee, K.S.; Cho, H.J.; Kim, Y.H.; Yang, H.K.; Kim, W.H.; Kang, G.H. Prognostic implications of tumor-infiltrating FoxP3+ regulatory T cells and CD8+ cytotoxic T cells in microsatellite-unstable gastric cancers. Hum. Pathol. 2014, 45, 285–293. [Google Scholar] [CrossRef]
  309. Ahn, G.O.; Tseng, D.; Liao, C.H.; Dorie, M.J.; Czechowicz, A.; Brown, J.M. Inhibition of Mac-1 (CD11b/CD18) enhances tumor response to radiation by reducing myeloid cell recruitment. Proc. Natl. Acad. Sci. USA 2010, 107, 8363–8368. [Google Scholar] [CrossRef] [Green Version]
  310. Soloviev, D.A.; Hazen, S.L.; Szpak, D.; Bledzka, K.M.; Ballantyne, C.M.; Plow, E.F.; Pluskota, E. Dual role of the leukocyte integrin alphaMbeta2 in angiogenesis. J. Immunol. 2014, 193, 4712–4721. [Google Scholar] [CrossRef] [Green Version]
  311. Sorrentino, C.; Miele, L.; Porta, A.; Pinto, A.; Morello, S. Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 2015, 6, 27478–27489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Schmaljohn, A.L.; Orlandi, C.; Lewis, G.K. Deciphering Fc-mediated Antiviral Antibody Functions in Animal Models. Front. Immunol. 2019, 10, 1602. [Google Scholar] [CrossRef] [PubMed]
  313. van Spriel, A.B.; van Ojik, H.H.; Bakker, A.; Jansen, M.J.; van de Winkel, J.G. Mac-1 (CD11b/CD18) is crucial for effective Fc receptor-mediated immunity to melanoma. Blood 2003, 101, 253–258. [Google Scholar] [CrossRef] [PubMed]
  314. Hsu, H.T.; Mace, E.M.; Carisey, A.F.; Viswanath, D.I.; Christakou, A.E.; Wiklund, M.; Onfelt, B.; Orange, J.S. NK cells converge lytic granules to promote cytotoxicity and prevent bystander killing. J. Cell Biol. 2016, 215, 875–889. [Google Scholar] [CrossRef]
  315. Kumar, S. Natural killer cell cytotoxicity and its regulation by inhibitory receptors. Immunology 2018, 154, 383–393. [Google Scholar] [CrossRef]
  316. Dustin, M.L. The immunological synapse. Cancer Immunol. Res. 2014, 2, 1023–1033. [Google Scholar] [CrossRef] [Green Version]
  317. Wurzer, H.; Hoffmann, C.; Al Absi, A.; Thomas, C. Actin Cytoskeleton Straddling the Immunological Synapse between Cytotoxic Lymphocytes and Cancer Cells. Cells 2019, 8, 463. [Google Scholar] [CrossRef] [Green Version]
  318. Fu, C.; Tong, C.; Wang, M.; Gao, Y.; Zhang, Y.; Lu, S.; Liang, S.; Dong, C.; Long, M. Determining beta2-integrin and intercellular adhesion molecule 1 binding kinetics in tumor cell adhesion to leukocytes and endothelial cells by a gas-driven micropipette assay. J. Biol. Chem. 2011, 286, 34777–34787. [Google Scholar] [CrossRef] [Green Version]
  319. Liang, S.; Slattery, M.J.; Dong, C. Shear stress and shear rate differentially affect the multi-step process of leukocyte-facilitated melanoma adhesion. Exp. Cell Res. 2005, 310, 282–292. [Google Scholar] [CrossRef] [Green Version]
  320. Hartmann, T.N.; Grabovsky, V.; Wang, W.; Desch, P.; Rubenzer, G.; Wollner, S.; Binsky, I.; Vallon-Eberhard, A.; Sapoznikov, A.; Burger, M.; et al. Circulating B-cell chronic lymphocytic leukemia cells display impaired migration to lymph nodes and bone marrow. Cancer Res. 2009, 69, 3121–3130. [Google Scholar] [CrossRef] [Green Version]
  321. Goldin, L.R.; McMaster, M.L.; Rotunno, M.; Herman, S.E.; Jones, K.; Zhu, B.; Boland, J.; Burdett, L.; Hicks, B.; Ravichandran, S.; et al. Whole exome sequencing in families with CLL detects a variant in Integrin beta 2 associated with disease susceptibility. Blood 2016, 128, 2261–2263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  322. Riches, J.C.; O’Donovan, C.J.; Kingdon, S.J.; McClanahan, F.; Clear, A.J.; Neuberg, D.S.; Werner, L.; Croce, C.M.; Ramsay, A.G.; Rassenti, L.Z.; et al. Trisomy 12 chronic lymphocytic leukemia cells exhibit upregulation of integrin signaling that is modulated by NOTCH1 mutations. Blood 2014, 123, 4101–4110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  323. Hutterer, E.; Asslaber, D.; Caldana, C.; Krenn, P.W.; Zucchetto, A.; Gattei, V.; Greil, R.; Hartmann, T.N. CD18 (ITGB2) expression in chronic lymphocytic leukaemia is regulated by DNA methylation-dependent and -independent mechanisms. Br. J. Haematol. 2015, 169, 286–289. [Google Scholar] [CrossRef] [PubMed]
  324. Till, K.J.; Harris, R.J.; Linford, A.; Spiller, D.G.; Zuzel, M.; Cawley, J.C. Cell motility in chronic lymphocytic leukemia: Defective Rap1 and alphaLbeta2 activation by chemokine. Cancer Res. 2008, 68, 8429–8436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Pye, D.S.; Rubio, I.; Pusch, R.; Lin, K.; Pettitt, A.R.; Till, K.J. Chemokine unresponsiveness of chronic lymphocytic leukemia cells results from impaired endosomal recycling of Rap1 and is associated with a distinctive type of immunological anergy. J. Immunol. 2013, 191, 1496–1504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  326. Packham, G.; Krysov, S.; Allen, A.; Savelyeva, N.; Steele, A.J.; Forconi, F.; Stevenson, F.K. The outcome of B-cell receptor signaling in chronic lymphocytic leukemia: Proliferation or anergy. Haematologica 2014, 99, 1138–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Montresor, A.; Bolomini-Vittori, M.; Simon, S.I.; Rigo, A.; Vinante, F.; Laudanna, C. Comparative analysis of normal versus CLL B-lymphocytes reveals patient-specific variability in signaling mechanisms controlling LFA-1 activation by chemokines. Cancer Res. 2009, 69, 9281–9290. [Google Scholar] [CrossRef] [Green Version]
  328. Benedicto, A.; Marquez, J.; Herrero, A.; Olaso, E.; Kolaczkowska, E.; Arteta, B. Decreased expression of the beta2 integrin on tumor cells is associated with a reduction in liver metastasis of colorectal cancer in mice. Bmc Cancer 2017, 17, 827. [Google Scholar] [CrossRef] [Green Version]
  329. Senbanjo, L.T.; Chellaiah, M.A. CD44: A Multifunctional Cell Surface Adhesion Receptor Is a Regulator of Progression and Metastasis of Cancer Cells. Front. Cell Dev. Biol. 2017, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  330. Wang, H.S.; Hung, Y.; Su, C.H.; Peng, S.T.; Guo, Y.J.; Lai, M.C.; Liu, C.Y.; Hsu, J.W. CD44 cross-linking induces integrin-mediated adhesion and transendothelial migration in breast cancer cell line by up-regulation of LFA-1 (alpha L beta2) and VLA-4 (alpha4beta1). Exp. Cell Res. 2005, 304, 116–126. [Google Scholar] [CrossRef]
  331. Yanguas, A.; Garasa, S.; Teijeira, A.; Auba, C.; Melero, I.; Rouzaut, A. ICAM-1-LFA-1 Dependent CD8+ T-Lymphocyte Aggregation in Tumor Tissue Prevents Recirculation to Draining Lymph Nodes. Front. Immunol. 2018, 9, 2084. [Google Scholar] [CrossRef] [PubMed]
  332. Zhang, M.; March, M.E.; Lane, W.S.; Long, E.O. A signaling network stimulated by beta2 integrin promotes the polarization of lytic granules in cytotoxic cells. Sci. Signal. 2014, 7, ra96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Anikeeva, N.; Somersalo, K.; Sims, T.N.; Thomas, V.K.; Dustin, M.L.; Sykulev, Y. Distinct role of lymphocyte function-associated antigen-1 in mediating effective cytolytic activity by cytotoxic T lymphocytes. Proc. Natl. Acad. Sci. USA 2005, 102, 6437–6442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Saed, G.M.; Fletcher, N.M.; Diamond, M.P.; Morris, R.T.; Gomez-Lopez, N.; Memaj, I. Novel expression of CD11b in epithelial ovarian cancer: Potential therapeutic target. Gynecol. Oncol. 2018, 148, 567–575. [Google Scholar] [CrossRef]
  335. Krueger, J.G.; Ochs, H.D.; Patel, P.; Gilkerson, E.; Guttman-Yassky, E.; Dummer, W. Effect of therapeutic integrin (CD11a) blockade with efalizumab on immune responses to model antigens in humans: Results of a randomized, single blind study. J. Investig. Dermatol. 2008, 128, 2615–2624. [Google Scholar] [CrossRef] [Green Version]
  336. Jelcic, I.; Jelcic, I.; Faigle, W.; Sospedra, M.; Martin, R. Immunology of progressive multifocal leukoencephalopathy. J. Neurovirol. 2015, 21, 614–622. [Google Scholar] [CrossRef]
  337. Major, E.O. Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies. Annu. Rev. Med. 2010, 61, 35–47. [Google Scholar] [CrossRef] [Green Version]
  338. Schwab, N.; Schneider-Hohendorf, T.; Melzer, N.; Cutter, G.; Wiendl, H. Natalizumab-associated PML: Challenges with incidence, resulting risk, and risk stratification. Neurology 2017, 88, 1197–1205. [Google Scholar] [CrossRef]
  339. Ho, P.R.; Koendgen, H.; Campbell, N.; Haddock, B.; Richman, S.; Chang, I. Risk of natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: A retrospective analysis of data from four clinical studies. Lancet Neurol. 2017, 16, 925–933. [Google Scholar] [CrossRef]
  340. Akaishi, T.; Nakashima, I. Efficiency of antibody therapy in demyelinating diseases. Int. Immunol. 2017, 29, 327–335. [Google Scholar] [CrossRef] [Green Version]
  341. Nelson, S.M.; Nguyen, T.M.; McDonald, J.W.; MacDonald, J.K. Natalizumab for induction of remission in Crohn’s disease. Cochrane Database Syst. Rev. 2018, 8, CD006097. [Google Scholar] [CrossRef] [PubMed]
  342. Potin, D.; Launay, M.; Monatlik, F.; Malabre, P.; Fabreguettes, M.; Fouquet, A.; Maillet, M.; Nicolai, E.; Dorgeret, L.; Chevallier, F.; et al. Discovery and development of 5-[(5S,9R)-9-(4-cyanophenyl)-3-(3,5-dichlorophenyl)-1-methyl-2,4-dioxo-1,3,7-tria zaspiro [4.4]non-7-yl-methyl]-3-thiophenecarboxylic acid (BMS-587101)--a small molecule antagonist of leukocyte function associated antigen-1. J. Med. Chem. 2006, 49, 6946–6949. [Google Scholar] [CrossRef] [PubMed]
  343. Suchard, S.J.; Stetsko, D.K.; Davis, P.M.; Skala, S.; Potin, D.; Launay, M.; Dhar, T.G.; Barrish, J.C.; Susulic, V.; Shuster, D.J.; et al. An LFA-1 (alphaLbeta2) small-molecule antagonist reduces inflammation and joint destruction in murine models of arthritis. J. Immunol. 2010, 184, 3917–3926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Celik, E.; Faridi, M.H.; Kumar, V.; Deep, S.; Moy, V.T.; Gupta, V. Agonist leukadherin-1 increases CD11b/CD18-dependent adhesion via membrane tethers. Biophys. J. 2013, 105, 2517–2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Jagarapu, J.; Kelchtermans, J.; Rong, M.; Chen, S.; Hehre, D.; Hummler, S.; Faridi, M.H.; Gupta, V.; Wu, S. Efficacy of Leukadherin-1 in the Prevention of Hyperoxia-Induced Lung Injury in Neonatal Rats. Am. J. Respir Cell Mol. Biol. 2015, 53, 793–801. [Google Scholar] [CrossRef] [Green Version]
  346. Khan, S.Q.; Guo, L.; Cimbaluk, D.J.; Elshabrawy, H.; Faridi, M.H.; Jolly, M.; George, J.F.; Agarwal, A.; Gupta, V. A Small Molecule beta2 Integrin Agonist Improves Chronic Kidney Allograft Survival by Reducing Leukocyte Recruitment and Accompanying Vasculopathy. Front. Med. (Lausanne) 2014, 1, 45. [Google Scholar] [CrossRef] [Green Version]
  347. Khan, S.Q.; Khan, I.; Gupta, V. CD11b Activity Modulates Pathogenesis of Lupus Nephritis. Front. Med. (Lausanne) 2018, 5, 52. [Google Scholar] [CrossRef] [Green Version]
  348. Kavanaugh, A.F.; Davis, L.S.; Jain, R.I.; Nichols, L.A.; Norris, S.H.; Lipsky, P.E. A phase I/II open label study of the safety and efficacy of an anti-ICAM-1 (intercellular adhesion molecule-1; CD54) monoclonal antibody in early rheumatoid arthritis. J. Rheumatol. 1996, 23, 1338–1344. [Google Scholar]
  349. Kavanaugh, A.F.; Davis, L.S.; Nichols, L.A.; Norris, S.H.; Rothlein, R.; Scharschmidt, L.A.; Lipsky, P.E. Treatment of refractory rheumatoid arthritis with a monoclonal antibody to intercellular adhesion molecule 1. Arthritis Rheum 1994, 37, 992–999. [Google Scholar] [CrossRef]
Figure 1. Representative members of integrins in vertebrates and their binding specificity.
Figure 1. Representative members of integrins in vertebrates and their binding specificity.
Ijms 21 01402 g001
Figure 2. Structure of β2 integrins.
Figure 2. Structure of β2 integrins.
Ijms 21 01402 g002
Figure 3. Conformational alterations determine the conformation and thereby the binding affinities of β2 integrins.
Figure 3. Conformational alterations determine the conformation and thereby the binding affinities of β2 integrins.
Ijms 21 01402 g003
Figure 4. Regulation of β2 integrin inside-out signaling. Activation of signaling adaptor proteins and β2 integrin-binding proteins in response to engagement of chemokine receptors and PSGL-1 is indicated by arrows.
Figure 4. Regulation of β2 integrin inside-out signaling. Activation of signaling adaptor proteins and β2 integrin-binding proteins in response to engagement of chemokine receptors and PSGL-1 is indicated by arrows.
Ijms 21 01402 g004
Figure 5. Schematic overview on the diverse functions of β2 integrins.
Figure 5. Schematic overview on the diverse functions of β2 integrins.
Ijms 21 01402 g005
Table 1. Role of β2 integrins in autoimmune diseases.
Table 1. Role of β2 integrins in autoimmune diseases.
Sub-UnitCell TypeSpecies 1Disease Model, Immune StateObservationsReference
CD18-hLADLAD patients suffer from intestinal colitis, periodontitis, Type 1 Diabetes, autoimmune cytopenia[166,252,253]
mLAD Chronic dermatitis and splenomegaly[177,179]
CD11aT cellmEAEPro- or anti-inflammatory functions, depending on the experimental setup[258,276,283]
hSystemic sclerosisExpression correlates with severity of disease[264,284]
hPsoriasisBlockade with Efalizumab induces T cell hyporesponsiveness and inhibits psoriasis pathogenesis[266]
hAutoimmune thrombocytopeniaHigh expression positively correlates with autoimmune thrombocytopenia pathogenesis[189]
hSLEExpression correlates with SLE severity[193]
hRAExpression correlates with RA pathogenesis[285,286]
mExpression is essential for CIA development[287]
CD11bDCmRAControls balance between Th17 and Treg via IL-6[255]
APCmPeripheral toleranceRequired for establishment of orally induced peripheral tolerance (suppresses IL-6/IL-17 induction)[249,288]
hRA, SLEPolymorphisms predispose for SLE and RA[193,276,289,290]
mSLEActivation suppresses autoimmunity[193]
DCmEAECD11b+ DC accumulate in CNS of MOG-immunized mice and present with tolerogenic phenotype (IL-10 and TGF-β secretion)[291]
T cellsmCD11b+ TC are required for the EAE development[269,292]
B cellmExperimental autoimmune hepatitisCD11b+ B cells suppress T cell response by inhibiting TCR signaling[189]
-hPsoriasisFrequency of CD11b+ cells in lesions correlates with MPO activity. CD11b expression in PMN/macrophages is elevated in pustular psoriasis.[293,294,295]
PMNmBullous pemphigoidCD11b is required for skin infiltration by PMN and inflammation development in anti-BP180 antibody-injected mice[275]
CD11cDCmAutoantibody productioncholesterol accumulation in DC contributes to autoimmune processes[296]
CD11d-h/mObesityCD11d expression is elevated in white adipose tissue of obese humans/mice[297]
1: h: human, m: mouse.
Table 2. The role of β2 integrins in tumor development.
Table 2. The role of β2 integrins in tumor development.
SubunitCell TypeSpeciesTumor ModelObservationsReference
CD18S100A8+ myeloid cellsmLewis lung carcinoma (LLC) and MC38 colon adenocarcinomaTumors growing in CD18hypo, but not in CD11b-deficient mice were more sensitive to irradiation in comparison to WT mice[309]
B cellhCLLCD18 variant (E630K) enhances CLL susceptibility[321,331]
CD11aNK, TCh-Establishment of an intercellular synapse with cancer cell and targeted release of granules depend on LFA-1[331,332,333]
B cellhCLLImpaired motility and accumulation in the blood due to defective Rap-1 GTPase signaling/diminished CD18 expression[320,324]
CD11aCD11bPMNmMelanomaICAM-1 expressing melanoma cells bind PMN via LFA-1/MAC-1, and thus are carried across the vasculature forming metastasis[319]
CD11bPMNmB16F10 Melanoma, RM1 prostate cancerCD11b−/− PMN fail to infiltrate tumor tissue and secrete VEGF needed for the neovascularization at lower extent[310]
mspontaneous intestinal adenomaCD11b−/− myeloid cell infiltrate tumor mass at low extent, associated with diminished tumor growth and impaired Wnt/β-catenin activity in the tumor[305]
mSquamous cell carcinoma xenograftsSystemic application of CD11b blocking antibody increased anti-tumor response after radiation[309]
hGastric cancerExtent of CD11b+ cell infiltration correlated with tumor size, venous invasion, lymph node metastasis, general metastasis stage and FoxP3+ cell infiltration[307]
hEpithelial ovarian cancer (EOC)de novo expression of MAC-1 on EOC cell lines[334]
mMelanomaMAC-1 is essential for antibody-mediated antitumor responses[313]
CD11cAPChGastric cancerCD11c+ cell tumor infiltration correlated with tumor size.[307]
CD11dMacro-phagem Upregulation of CD11d/CD18 surface expression by myeloid cells is associated with their accumulation at the inflammation site and chronification of inflammation[67]

Share and Cite

MDPI and ACS Style

Bednarczyk, M.; Stege, H.; Grabbe, S.; Bros, M. β2 Integrins—Multi-Functional Leukocyte Receptors in Health and Disease. Int. J. Mol. Sci. 2020, 21, 1402. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041402

AMA Style

Bednarczyk M, Stege H, Grabbe S, Bros M. β2 Integrins—Multi-Functional Leukocyte Receptors in Health and Disease. International Journal of Molecular Sciences. 2020; 21(4):1402. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041402

Chicago/Turabian Style

Bednarczyk, Monika, Henner Stege, Stephan Grabbe, and Matthias Bros. 2020. "β2 Integrins—Multi-Functional Leukocyte Receptors in Health and Disease" International Journal of Molecular Sciences 21, no. 4: 1402. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041402

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop