Abstract
Pattern recognition receptors (PRRs) are germline-encoded sensors best characterized for their critical role in host defense. However, there is accumulating evidence that organ transplantation induces the release or display of molecular patterns of cellular injury and death that trigger PRR-mediated inflammatory responses. There are also new insights that indicate PRRs are able to distinguish between self and non-self, suggesting the existence of non-clonal mechanisms of allorecognition. Collectively, these reports have spurred considerable interest into whether PRRs or their ligands can be targeted to promote transplant survival. This review examines the mounting evidence that PRRs play in transplant-mediated inflammation. Given the large number of PRRs, we will focus on members from four families: the complement system, toll-like receptors, the formylated peptide receptor, and scavenger receptors through examining reports of their activity in experimental models of cellular and solid organ transplantation as well as in the clinical setting.
The Complement System
The complement cascade is a component of the immune system that is evolutionarily ancient, and is involved in rapidly clearing pathogens and debris from the circulation [1]. Over a period of time, it has evolved into a key component that bridges the innate and adaptive immune responses. It is a family of over 60 proteins and its components serve a multitude of roles, including being an anaphylatoxin (i.e., C3a, C5a) for leukocytes, serving as an opsonin to facilitate phagocytes to clear debris (i.e. C3b), and participating in a membrane-attack complex to lyse bacteria (i.e. C5b-9), which can assemble on bacteria (such as Neisseria) and lyse it. In the absence of regulatory proteins, uncontrolled activation of the system can also result in disruption of host cells, resulting in immune-mediated tissue damage.
The complement system has a long-standing role in ischemia-reperfusion injury (IRI) of multiple organ systems [2–4]. Different components of the cascade have been implicated, including members of the classical, alternative [5, 6], and lectin pathways [7, 8]. Certain complement proteins serve as pattern recognition molecules, notably C1q, which is a component of the classical pathway of complement activation [9–12]. Additionally, components of the lectin pathway of activation increasingly have come to be appreciated as pattern recognition molecules [13, 14]. Multiple complement proteins also provide a cross-talk pathway with other pattern recognition molecules, notably with Toll-like receptors [15–17]. As a result of these diverse roles, the complement system increasingly is seen as an important mediator of inflammatory responses to transplanted organs [18–24] and has been a longstanding therapeutic target of interest [25–30].
The central component of the complement cascade is the protein C3, which is the most abundant protein in the circulation after albumin and α2-macroglobulin [31]. While the primary source of C3 is the liver, extrahepatic production of C3 has been found to be involved in outcomes of solid organ transplantation. For example, the local production of C3 in the kidney has been shown to be important in kidney ischemia-reperfusion injury (IRI), as well as generation of an alloimmune response [32]. Renal IRI also has been known to amplify the humoral immune response [33]. Anaphylatoxins such as C3a and C5a, generated from immune cells themselves, also have the ability to modulate alloimmune responses [20, 21, 34–36]. The primary/traditional mechanism by which complement contributes to tissue injury is through the formation of the membrane attack complex (MAC, C5b-9). Additionally, sublytic MAC formation can activate the NLRP3 inflammasome, as well as intracellular NF-κB activation [37]. In a heart transplant model, this effect was also associated with activation of allogeneic CD4 T cells [38]. Finally, while IRI may be driven by activators, the contribution of the relative deficiency/downregulation of the regulators [39] also has become increasingly examined. This review focuses on the triggers of the cascade that also serve as pattern-recognition molecules and have a known association/mechanistic link with solid organ transplant outcomes.
C1q
C1q is a known pattern-recognition molecule that activates the classical pathway of the complement cascade by binding to the Fc region of antibodies (IgM, IgG) and on membrane receptors of cells, such as cC1qR/CR (calreticulin) and gC1qR/p33 [40] [41]*), and annexin A2/A5 on apoptotic cells [42]. C1q deficiency is associated with autoimmunity [40, 43], which has been shown to be mitigated by bone marrow transplantation in pre-clinical models [44, 45]. C1q can modulate CD8 responses by controlling cellular metabolism [46]. It can bind to apoptotic cells and suppress macrophage and dendritic-cell mediated Th1 and Th17 polarization [47]. It can also limit inflammasome activity in macrophages [48]. While there is a strong fundamental basis for C1q to facilitate immunoregulation, its role in solid organ transplantation is only beginning to be elucidated. In a cardiac allograft mouse model (BALB/c > B6), Csencstis and colleagues showed that C1q−/− recipients had worse acute cellular rejection compared to wild-type recipients as demonstrated by increased immune cell infiltrate in the myocardium, hemorrhage, and myocyte necrosis [11]. C1q−/− mice had significantly higher proportions of %CD19+ B cells, lower %CD4 T cells, and no significant differences in %CD8 T cells in the spleen, as well as no differences in donor-reactive T cell responses. Interestingly, C1q−/− mice had an accelerated isotype switch of donor reactive alloantibodies (IgG), IgG deposition in the allograft, and C3d deposition in the allografts, despite no C4d deposition. These observations suggest that although C1q activates the classical pathway, allograft injury may be driven by components of the lectin and alternative pathways. Whether antibody-mediated rejection is contributing to the allograft injury despite the absence of activating the classical pathway, remains to be elucidated.
Apart from the studies on C1q in autoimmunity, understanding how it facilitates tolerance has been primarily studied using a mouse minor histocompatibility (HY) model of skin graft rejection. Using this model, Baruah and colleagues demonstrated that female C1qa−/− mice had a higher IFN-γ producing CD8+ Teff population, and a higher IL-10 producing CD8+Teff population, compared to wild-type females, when transplanted with skin from male mice. C1q was necessary to facilitate tolerance by intranasally administered HY peptide [49]. Subsequent work from the same group [50] has suggested that this effect is specific to C1q, C3, and C4 (i.e. early components of the classical pathway), and demonstrated the redundancy of C5 in this model. C3-deficiency in this model resulted in impaired iNOS upregulation, leading to decreased T-regulatory cell induction. While these experiments were not specifically shown in C1qa−/− mice, the authors suggested that the classical pathway, via C3, played an important role in intranasal peptide-mediated tolerance induction via the T-regulatory cell-dendritic cell tolerogenic loop. More recently, C1q has been shown to activate Wnt signaling and promote skeletal muscle aging [51]; whether this role is applicable in senescent allografts remains to be evaluated.
Pentraxins, ficolins and collectins
In addition to C1q, other notable pattern recognition molecules in the complement system that are relevant to solid organ transplantation are pentraxins, ficolins and collectins [52]. Many of these proteins serve as triggers for different pathways of the complement cascade. A majority of the data for ficolins exists primarily in humans [53]. These are soluble oligomeric proteins containing trimeric collagen-like regions linked to fibrinogen-related domains (FReDs) that serve as pattern-recognition molecules for pathogens and dying cells, and activate the complement system. For example, they can bind to lipopolysaccharide, components of cell walls (i.e., D-fucose), and pentraxins to activate the complement system via the classical and lectin pathways. While changes in their levels have been associated with worse clinical outcomes after renal transplantation [54, 55], a majority of the experimental models have been conducted to study the role of pentraxins and collectins, not ficolins.
Pentraxins are a family of soluble proteins that can bind to multiple pathogens, as well as host surfaces (e.g., C1q, extracellular matrix proteins, leukocytes, dying cells), to activate the classical and lectin pathway [56]. Based on the length of their primary structure, they are classified into short- and long-chain pentraxins. C-reactive protein (CRP) and serum amyloid P (SAP) are the prototypical short-chain pentraxins, while long pentraxin-3 (PTX3, previously referred to as TSG-14, TNF-stimulated gene 14), is the prototypical long-chain pentraxin [57]. CRP and SAA are primarily synthesized by the liver, and their synthesis is augmented in the setting of pro-inflammatory stimuli, notably IL-6, resulting in their serving as acute phase reactants. PTX3, on the other hand, is produced by multiple sources, including endothelial cells, fibroblasts, lungs, heart, ovaries, thymus, and skin. Its production also is upregulated by pro-inflammatory stimuli such as TNFα and IL-1β [58]. CRP and PTX3 have been studied in the context of allograft injury, with a considerable focus on PTX3, particularly due to its interplay with C1q, among other immunomodulatory effects.
CRP is a fluid-phase pattern-recognition molecule that can bind to pathogens, as well as ligands on injured cells/tissues [59]. As an acute-phase reactant, it has a baseline level of < 5 mcg/mL in human blood, which can increase to 500 mcg/mL in the setting of an injury/infection. CRP has a cyclic pentameric structure, which facilitates its role as a pattern-recognition molecule. On one surface, its five calcium-dependent binding sites allow it to bind to its target. On its opposite surface, it is able to bind to C1q to activate the classical pathway of complement and (FcγRs) on phagocytic cells [59, 60]. Interestingly, this FcγR-facilitated interaction with macrophages can result in the synthesis and deposition of C3 at the site of injury [61]. Those investigators used human CRP transgenic mice in combination with an FcγR- or C3-deficiency in a vascular injury model to demonstrate that the neointimal thickening associated with vascular injury was dependent on FcγR, and is complement (i.e. C3)-dependent.
Interestingly, although C1q binds to CRP, and C1q itself is immunoregulatory, CRP appears to worsen tissue damage in experimental models of renal and hepatic IRI. In a series of two manuscripts, Pegues and colleagues showed that CRP worsened renal IRI by shifting the balance of kidney-infiltrating myeloid-derived suppressor cells (MDSCs) toward a suppressive phenotype [62]. Using human CRP transgenic mice in a model of warm IRI, they showed that compared to wild-type mice, these transgenic mice had worsened physiological and histological markers of renal injury and increased FcγRI expression. There were alterations in expression of macrophage activation markers; however, the specific mechanism by which CRP worsened renal IRI remained unclear. A followup study by the same group demonstrated that CRP-transgenic mice had a higher proportion of g-MDSCs (Gr1+CD11b+Ly6g+Ly6clow) cells in their injured kidneys, compared to wild-type mice subjected to IRI. Additionally, the depletion of these g-MDSCs using an anti-Gr1 (depleting) antibody, reduced urine albumin levels at 24 h after renal IRI in the CRP-transgenic mice. Thus, one potential mechanism of how CRP worsens IRI has been deciphered; however, how these MDSC populations are altered (i.e., receptors involved), what other factors are playing a role in CRP-mediated IRI, and what would be the optimal targets for intervention remain unanswered.
Two other studies also warrant mention. By mating transgenic (Tg)-CRP mice to two autophagy reporter mouse lines, Tg-GFP-LC3 mice (LC3) and Tg-RFP-GFP-LC3 mice (RG-LC3) respectively, Bian and colleagues reported that CRP impaired autophagic flux in vivo (measured by LC3 II/I and p62 levels by immunoblotting, GFP-LC3 punctae post-IRI by immunohistochemistry), ex vivo and in vitro in a warm renal IRI. CRP also impaired the dissociation of beclin-1 to bcl-2, which is required for autophagy, which was rescued by rapamycin [63]. Interestingly, Thiele and colleagues showed that a conformational change in pentameric CRP to isoforms expressing pro-inflammatory neo-epitopes was needed for inducing IRI in rat kidneys. They proposed that circulating pCRP could bind to activated biomembranes in the microcirculation of the injured tissue and be converted to bioactive pCRP (expressing neoepitopes), dissociate, and form monomeric CRP (mCRP) [64]. Those structurally altered CRP isoforms facilitated leukocyte recruitment by increasing leukocyte rolling and adhesion to endothelial cells (determined using intravital microscopy) and ROS formation in leukocytes (by activating NAPDH oxidase enzyme complex). Tested therapies for reducing that injury in their pre-clinical model system included 1,6-bis(phosphocholine)-hexane (1,6-bisPC), which stabilized pCRP and prevented the conformational changes that expose pro-inflammatory neo-epitopes, and nystatin, which reduced ROS formation in leukocytes by blocking lipid-raft dependent pathways. CRP activation, with IgM and C3 deposition has also been observed in rat hepatic IRI model, suggesting that the pro-inflammatory role of CRP in IRI may be applicable in multiple organ systems [65]. However, the specifics in each organ solid organ transplant system remain to be explored, and current knowledge is largely built upon what has been shown in renal transplant literature.
Long pentraxins are a family of proteins that have an unrelated N-terminal coupled to a pentraxin-like C terminal [8, 66]. The prototype long pentraxin, PTX3, acts as an acute-phase reactant, whose synthesis is increased by TNFα and IL-1β. Unlike short pentraxins (i.e., CRP), functionally relevant PTX3 has extrahepatic sources, including the heart, lungs, and bone marrow [8]. This local synthesis results in unique phenotypes in the context of IRI and transplantation, many of which have their basis in the biology of PTX3. Specifically, PTX3 and C1q can reciprocally bind to each other, and have different functions in the clearance of apoptotic cells. For example, while C1q is responsible for binding to apoptotic cells and facilitating their clearance by macrophages, soluble PTX3 removes bound C1q from apoptotic cells, leading to impaired complement activation on dying cells, and reduced C1q-mediated phagocytosis [67]. This would suggest PTX3 impairs recognition of self-antigens, promoting autoimmunity. If this concept was then extrapolated to IRI in the allograft tissue, PTX3-deficiency would increase the risk of alloantigen exposure, increasing the risk of allograft rejection. PTX3 also can interact with FcγRIII to facilitate clearance of apoptotic debris [68]. In the absence of this interaction, other pattern recognition molecules of the innate immune system infiltrate into the injured tissue, contributing to injury.
The concept that PTX3 limits tissue injury has been observed in models of heart and renal IRI and transplantation. While it was initially demonstrated that exogenously administered PTX3 limited cardiac injury in acute myocardial infarction [69], Zhu and colleagues more recently showed that an exogenous administration of PTX3 reduced myocardial injury after heart transplantation, and regulated the expansion of γδ T cells, the major source of IL-17A [70]. Conversely, a neutralizing antibody against PTX3 worsened cardiomyocyte apoptosis and increased the recruitment of neutrophils and macrophages. Interestingly, the local source of PTX3 that reduces cellular injury has become of increasing interest. Shimizu and colleagues used chimeric mice in a model of myocardial IRI, and found that PTX3 needed to be generated from bone-marrow derived cells, not tissue-resident cardiac cells, to attenuate myocardial injury via attenuating neutrophil infiltration, ROS generation and proinflammatory cytokine production (i.e., IL-6) [71].
A similar phenotype has also been observed in a renal hilar clamp model (warm IRI), wherein PTX3 was found to attenuate IRI-induced fibrosis via IL-6-STAT3 axis [72]. A different group of investigators demonstrated that the reno-protective effect of PTX3 may occur through tissue-resident (intra-renal) CD45+CD11c+cells and CD45+CD11b+ cells (and not the CD45− cells) by regulating neutrophil adhesion and transmigration, attenuating TNFα and IL-6 release, and tubular necrosis [73]. Those manuscripts suggested that while the immunoregulatory effects of PTX3 may not be restricted to a single organ, the source of the protective PTX3 may be unique, and recipient versus donor PTX3 sources (and levels) would need to be considered when evaluating how best to augment PTX3 levels at the site of injury. This is especially important in the context of lung transplantation; where experimental data are lacking, and the main known finding in humans is that elevated long PTX3 levels are associated with primary graft dysfunction, a consequence of lung IRI and a form of acute lung injury, in lung transplant recipients with pulmonary fibrosis. The source of PTX3 and whether it is protective or deleterious in the context of lung transplantation is an area of active investigation of our laboratory.
Similar to pentraxins, collectins also are pattern-recognition molecules used by the complement system, their name representing collagen-like lectins [74]. Of these collectins, the most commonly known lectin is mannose-binding lectin (MBL), which is primarily produced by hepatocytes. However, there are a number of collectins that are locally synthesized, and are increasingly seen to be pattern-recognition molecules that influence outcomes in organ transplantation, notably collectin-11 [75]. Surfactant proteins (i.e., SP-A and SP-D) also are collectins that serve as pattern-recognition receptors. They contain both collagen-like sequences at the N terminus and calcium-dependent lectin domains at the C terminus, and provide immunomodulatory effects specific to the lung [76–78]. Studies in humans revealed that low levels of SP-A in the donor lung prior to implantation were associated with lower survival, and that phenotype may be driven by donor SP-A2 polymorphisms [79, 80]. Those studies suggested that the effects of decreased SP-A in the lung largely affected outcomes in the first year after transplantation [80, 81]. However, given that most experimental research on the role of surfactant proteins in organ transplantation has occurred over 10 years ago [82, 83], this discussion is focused on advances in the understanding of the role of MBL and collectin-11.
Mannose-binding lectin (MBL) and MBL-associated serine proteases (MASP)
MBL is a protein consisting of O-glycosylated polypeptide chains, the basic subunit of which is a trimer, that can form a series of higher oligomers. These multimers of the triple subunit can form a three-dimensional structure similar to C1q, thus facilitating pattern recognition [74, 84]. Among the four regions on each polypeptide, the collagen-like domain interacts with enzymes known as MBL-associated serine proteases (MASPs), and can also bind to receptors such as cC1qR (calreticulin), CR1, and C1qRp on phagocytes [85]. This association with MASP allows MBL to act as an opsonin and activate the lectin pathway of complement. Additionally, the interaction with phagocytes can influence the cellular immune response, similar to its interaction with other pattern-recognition molecules such as pentraxins. While the liver is the primary source of MBL [86], it is also detected in other body fluids, including bronchoalveolar lavage fluid. and urine. Genetic polymorphisms considerably influence its levels in the circulation [87]. As a pattern-recognition molecule, MBL appears to regulate tissue injury in experimental models of heart, kidney, and gut IRI [88]. Specifically, mice and rats lacking MBL are protected from myocardial IRI [89–91]. One proposed mechanism for MBL-mediated IRI is that the injured endothelium binds to MBL and IgM, which results in complement activation, and can set up a cascade of events that results in NLRP3 inflammasome assembly and IL1β release [92]. In the context of murine cardiac transplantation, recipient MBL-initiated complement activation contributed to an increased proportion of donor-reactive, IFNγ-producing spleen cells, which could be targeted in the perioperative period (day 0 and 1 post-transplantation) using C1-inhibitor therapy in combination with CTLA-Ig [93].
The role of MBL-initiated allograft injury, and targeting it with C1-inhibitor therapy, also has been studied in renal transplantation. Specifically, in a swine model of renal IRI, C1-inhibitor therapy: (1) decreased tubular damage, including tubular epithelial cell death, (2) decreased peritubular capillary and glomerular complement deposition (i.e. C4d, C5b-9), and (3) decreased the number of kidney-infiltrating CD163+ and T effector cells (CD4+ and CD8+) [94, 95]). However, there are a couple of caveats to consider in MBL-initiated allograft injury and targeting it. First, the effects of MBL may be complement-independent. Van der Pol and colleagues demonstrated that exposure of tubular epithelial cells to circulating MBL resulted in rapid internalization of MBL and tubular epithelial cell death that could not be prevented in spite of circulating C3 levels being depleted by using cobra-venom factor [95]. Second, the effect of C1-inhibitor therapy extendd beyond simply inhibiting the classical and lectin pathways of complement activation [96–98]
Among the MBL-associated serine proteases, MASP2 is the main enzyme that has been demonstrated to modify outcomes in experimental solid organ transplantation. MASP2 occurs as a single-chain polypeptide synthesized by the liver. It has domains analogous to C1r, C1s, MASP-1, and MASP-3, and undergoes calcium-dependent dimerization. It is generated as a pro-enzyme which is activated by cleavage to form a quaternary structure comprised of α- and β-chains linked by a disulfide bond. It can bind to collagen-like domains of MBL and certain ficolins and collectins, which serve as pattern-recognition molecules of the lectin pathway. As a result of these interactions, it circulates bound to these proteins, but becomes activated when it is concentrated on a target surface to cleave C2 and C4 to generate a lectin pathway convertase (C4bC2a) and activate the complement cascade. MASP2 can be either autoactivated in these settings or cleaved by MASP-1 [99]. Notably, MASP2 is the only protease of the lectin pathway that can cleave C4; thus making it critical for lectin pathway activation [100]. In addition, it can activate the cascade by bypassing C4 (due to its interactions with MASP-1 and C2 [101], and can promote clotting. Hence, this enzyme plays an important role in lectin-pathway-mediated tissue injury.
The effects of MASP-2 on tissue injury have been demonstrated in renal, cardiac and gastrointestinal IRI models. In the process of understanding whether the role of MASP2 is specifically through C4, Schwaeble and colleagues showed that MASP2-deficient mice had smaller infarct volumes than wild-type mice undergoing transient myocardial IRI. Both genetic MASP2 deficiency and pharmacologic abrogation of MASP2 activation, using a murine-specific MASP-2 inhibitor, AbyD 04211, also mitigated gastrointestinal IRI [101]. This approach was subsequently validated by Asgari and colleagues in a model of renal IRI in the setting of syngeneic renal transplantation, where the activity of MASP2 via its C4 bypass mechanism was identified as a contributor to delayed graft function [102].
Interestingly, MASP-2 does not appear to be an acute phase reactant. In other words, precise substrates need to be made available locally, and/or be deposited on an activated surface for MASP-2 to activate both C4-dependent and -independent mechanisms of complement cascade activation; and this activation can be therapeutically harnessed in experimental models to reduce IRI. Along those lines, Farrar and colleagues showed that L-fucose is exposed on renal tubular epithelial cells after IRI, and thats ligand binds to collectin-11 (CL-11), whose synthesis is rapidly increased in the kidney in the setting of IRI [75]. L-fucose has a known motif for binding to MASP-1, MASP-2 and MASP-3. The L-fucose-CL-11 interaction triggered MASP-2 activation, local complement activation, and tissue injury, and could be targeted using sugar-moiety specific (i.e., fucosidase) treatment, as demonstrated in vitro.
Toll-like receptors (TLR)
TLRs are a group of PRRs that are broadly expressed on immune cells. Their expression and function has been characterized mainly on innate immune cells such as neutrophils, monocytes, macrophages, dendritic cells, and natural killer cells [103], although they have been found to play important roles in B and T lymphocytes [104]. To date, 11 human and 13 mouse TLRs have been identified that recognize distinct pathogen-derived or endogenous ligands, all signaling through MyD88, except for TLR3. They were originally identified on innate immune cells by their ability to drive inflammatory cytokine expression and promote antigen presentation following engagement with pathogen associated molecular patterns [105, 106]. However, later work by Matzinger and colleagues [107] demonstrated that TLRs also could sense sterile cellular injury through recognition of damage-associated molecular patterns (DAMPs), which structurally resemble PAMPs (Table 2). Upon engagement, TLRs signaling is largely differentiated by the recruitment of two adaptor molecules, MyD88 and TRIF. MyD88-dependent signaling leads to the activation of the IKK complex, which results in the activation of MAP kinases and NF-κB. MyD88-independent pathways signal through TRIF, which leads to the activation of TBK1 and RIPK1 kinases. TBK1 pathways lead to IRF3 phosphorylation and Interferon type I production. RIPK1 pathways activate TAK1, leading to NFκB transcription [108]. TLR4 is the only TLR that utilizes both MyD88 and TRIF signaling pathways [109].
Table 2.
Pattern Recognition Molecule | Ligand | Target Organ | Injury |
---|---|---|---|
TLR2 | HMGB1 Heat shock protein Hyaluronan | Skin | -Shortens allograft survival time via MyD88 signaling and enhancing Th1 responses [110] -Abrogates the effect of costimulatory blockade by preventing alloreactive CD8+ T cell apoptosis [123] |
Heart | -worsens IRI [117] | ||
TLR4 | HMGB1 Heat shock protein Hyaluronan Fibronectin Heparan sulfate | Skin | - Abrogates the effect of costimulatory blockade by preventing alloreactive CD8+ T cell apoptosis [123] |
Kidney | - TLR4 signaling by tubular epithelial cells worsens IRI [121] | ||
Heart | - Worsens myocardial IRI [116] | ||
Lung | - worsens IRI [119, 120] | ||
Liver | - Worsens IRI via IRF3-dependent pathway [118] | ||
TLR9 | Mitochondrial DNA HMGB1 | Liver | - worsens IRI [200] |
Kidney | - worsens IRI [201] | ||
Heart | - worsens IRI [202] | ||
Lung | -- Promotes NET formation in primary graft dysfunction post-IRI [154] | ||
FPR1 | Formylated Peptides | Liver | - Promotes necrotaxis and exacerbates IRI [155] |
Lung | - Promotes airway neutrophilia and exacerbates IRI [148] | ||
Heart | - Increases inflammation, cardiomyocyte apoptosis and ventricular remodeling post IRI [203] |
Abbreviations: HMGB1: High Mobility Group Box 1; IRI: ischemia-reperfusion injury; NET: neutrophil extracellular traps; TLR: Toll-like receptor
The importance of TLR expression in a transplant setting was first recognized by the seminal work by Goldstein and Lakkis who demonstrated that MyD88 inhibits indefinite allograft survival in a mouse model of minor antigen-mismatch skin transplantation [110]. They reasoned this was due to reduced numbers of donor DCs trafficking into draining lymph nodes resulting in impaired alloantigen-specific T cell generation. Notably, TLR-signaling dependent DC trafficking was later observed in a complete major and minor MHC mismatched model of mouse skin transplantation, although both MyD88 and Trif deficiency were required to extend allograft survival in this setting [111]. Later studies have largely focused on the contribution of TLR2 and TLR4 to graft survival given several DAMPs associated with solid organ injury are recognized by both TLRs. These include heat shock proteins [112, 113] and hyaluronic acid (HA) [114] [115]. Interestingly, TLR4 is a very highly cited mediator of heart [116, 117], liver [118], lung [119, 120], and kidney [121] IRI suggesting it may play a dominant role in perioperative transplant injury although it is unclear as to the identity of the cognate ligand(s) in many of these reports. However, in generating downstream adaptive immune responses to transplanted organs both TLR2 and 4 are likely both required [110]. In a model of mouse orthotopic lung transplantation, recipient double deficiency in TLR2 and 4 were necessary to inhibit low molecular weight hyaluronic acid -mediated acute rejection [122]. The same study also showed that generation of alloreactive effector T cells was sharply reduced in TLR2−/−TLR4−/− recipients.
Additional insight into the role of TLRs in graft survival have been gained through studying the effects of pathogen associated molecular pattern administration to experimental transplant models pointing to the possible underlying mechanisms of infection-associated allograft rejection. For example, the co-injection of Pam3Cys, LPS, or bacterial CpG DNA oligonucleotides, respective ligands for TLR2, TLR4 and TLR9, prevented co-stimulatory blockade-mediated allograft survival by impairing the deletion of alloreactive CD8+ T cells in a mouse model of skin transplantation [123]. Several reports have also suggested that TLR-mediated inflammatory responses prevent tolerance through inhibiting the suppression, recruitment or stability of Foxp3+ CD4+ T regulatory (Treg) cells. Medzhitov and colleagues first demonstrated that TLR-driven IL-6 expression by dendritic cells inhibits Treg mediate-suppression of effector T cells [124]. Interestingly, CpG DNA-mediated IL-6 and IL-17 was later shown to prevent anti-CD154-mediated conversion of conventional T cells into Tregs as well as preventing mouse heart allograft tolerance [125]. However, the role of IL-6 remains controversial as others have shown in multiple experimental transplant models the existence of IL-6 independent mechanisms that impair Treg suppressive capacity in response to CpG DNA stimulation [126]. Although these effects are likely mediated by TLR9, intracellular PRRs such as STING have been shown to recognize double stranded DNA, making still unclear if targeting TLR9 would be an effective strategy at promoting Treg-mediated effects [127]. It is also likely that other TLR ligands play a role on Treg stability or recruitment. In a mouse islet transplantation model, TLR4 deficiency led to augmented rapamycin-induced Treg expansion and prolonged allograft survival [128]. Additionally, Hsieh and colleagues have demonstrated that silencing TLR4-MyD88 signaling pathway in femoral bone transplants promoted Treg expansion along with allograft survival [129].
TLR reactivity and expression patterns have also been linked to clinical outcomes after transplantation. For example, by measuring TLR reactivity in reporter cells line exposed to blood samples from liver transplant recipients, Sosa et al recently demonstrated that a positive TLR4 and 9 reactivity to the post-reperfusion samples was a major risk factor for IRI associated with orthotopic liver transplantation, especially when coupled to a negative reactivity to pre-reperfusion samples [130]. Higher levels of TLR4 and TLR2 have been detected in patients with developing kidney graft dysfunction [131] and liver cellular rejection [132] when compared to recipients with stable transplants. Moreover, higher TLR4 expression and increased TNFα and IL-6 responses to LPS in monocytes from patients before liver transplantation also has been associated with increased risk of graft rejection [133]. Additionally, increased survival has been observed in lung recipients carrying the hyporesponsive TLR4 polymorphisms Asp299Gly or Thr399Ile [134]. Altogether, those findings suggest that either profiling TLR expression or assessing genetic TLR polymorphisms may have potential applications in predicting the risk for allograft rejection.
Although targeting TLRs may be advantageous to promote allograft survival, new work has also illuminated their importance in maintaining suppressive cell populations. For example, TLR2 and 4 stimulation has been shown to promote Treg numbers and suppressive function in the intestine following infection with Fusobacterium nucleatum [135]. Repeated administration of LPS was also shown to drive Treg expansion and prevent experimentally-induced allergic asthma and autoimmune diabetes [136]. Interestingly, TLR4 drives the expansion of CD11b+Griint(Ly6Gint)F4/80+ cells, which display suppressive activity similar to myeloid derived suppressor cells (MDSCs) [137]. Several reports have demonstrated a critical role of MDSCs in promoting kidney, heart and lung transplantation tolerance [138, 139].
Finally, TLR expression by non-immune cells [140] may be helpful in protecting tissue from injury. Liang and colleagues, using a mouse model of bleomycin-induced lung injury, showed that TLR4 expression on lung epithelial cells is protective against tissue injury through stimulating NF-κβ dependent-survival pathways [141]. A subsequent study by the same group demonstrated that, upon engagement with high molecular weight HA, TLR4 signaling promoted type II alveolar epithelial cell proliferation and renewal. Thus, although TLR engagement led to immune cell activation, it may also be important in protecting parenchymal tissues from death and fibrosis [142]. Taken together, those reports suggest that TLRs are central players in linking innate and adaptive immune responses against transplanted organs. Further studies on the role of TLR-mediated regulatory mechanisms will be needed to consider the use of this approach to promote transplant tolerance.
FPR1/TLR9
FPR1 is a seven transmembrane G-protein–coupled receptor for the N-formyl methionine peptides, products of bacterial peptide synthesis that are well-established neutrophil chemo-attractants [143]. FPR1 also stimulates ROS generation, phagocytosis and the degranulation of oxidants and proteases from neutrophils [144]. FPR1 signaling is mediated by PI3Kγ activation that leads to calcium flux that drives neutrophil cytoskeletal reorganization and respiratory burst activity [144]. TLR9 is an intracellular receptor highly expressed in the ER of resting immune cells such as plasmacytoid dendritic cells, macrophages and B cells. Following engagement with hypomethylated CpG DNA motifs, TLR9 relocates to lysosomes where it co-localizes with MyD88 to drive signaling that promotes the expression of pro-inflammatory cytokines, including type I interferons IL-6, TNF-α, and IL-12 [145].
Although FPR1 and TLR9 play critical roles in host defense response to bacterial infection, there is increasing recognition that they play similar and complementary roles in the recognition of mitochondria released following necrotic cellular injury [146]. Mitochondria, likely due to their endosymbiotic origin, express N-formylated peptides and encode hypomethylated CpG DNA motifs in their genome that can be recognized by FPR1 and TLR9, respectively. Hauser and colleagues were the first to demonstrate the pathological consequences of mitochondria-derived CpG DNA (mt-DNA) and N-formylated peptide release in patients suffering from non-infectious traumatic crush injury leading to severe inflammatory respiratory syndrome (SIRS) [147]. The group observed high levels of circulating mt-DNA in SIRS patients and that mitochondrial damage associated molecular patterns (mt-DAMPs) stimulated neutrophil calcium flux and FPR1-dependent chemotaxis along with severe solid organ damage in rodents. Recently, there are several reports of high levels of circulating plasma mt-DNA in recipients of lungs [148] and kidneys [149] with perioperative graft dysfunction. Although, it has been presumed that mt-DAMPs originate from the transplanted organ, a study using an orthotopic lung transplant model in which donor lungs encoded a mitochondrial-targeted fluorescent protein showed nearly 25% of mitochondria released following IRI originated from the recipient [148]. One possible explanation for that observation is that neutrophils can release mt-DNA through the generation of neutrophil extracellular traps [150, 151]. Interestingly, NETosis has been shown to contribute to post-graft dysfunction in clinical lung transplantation and in models of lung transplantation [152, 153]. However, it remains to be determined whether neutrophil-derived mt-DNA exacerbates graft damage, as it recently has been demonstrated that mt-DNA itself can stimulate TLR9-dependent NETosis [154].
Approaches to inhibit graft injury by mitigating the effects of mt-DAMPs have focused on targeting neutrophil trafficking. In a mouse model of warm liver IRI, Honda and colleagues utilized intravital imaging to reveal that FPR1 promoted short-range neutrophil chemotaxis into areas of hepatic necrosis [155]. The group also pre-treated mouse livers with Cyclosporine H, a pharmacological inhibitor of FPR1, and demonstrated reduced neutrophil recruitment into nonperfused hepatic tissue and attenuated IRI. Consistent with those observations, in a orthotopic mouse lung transplant model, FPR1 was shown to direct neutrophil trafficking into airspaces, resulting in exacerbation of IRI. Additionally, graft-infiltrating FPR1−/− neutrophils were found to have engulfed fewer donor-derived Mt-DAMPs and produced lower levels of reactive oxygenation species [148]. Nevertheless, further work will be needed to determine if targeting PRRs that recognize mt-DAMPs is viable strategy to prevent graft injury. Additionally, it remains unknown whether either FPR1 or TLR9 contributes to adaptive immune responses against allografts.
Scavenger Receptors
The innate immune system recognizes molecular patterns that allow for the differentiation between healthy and dying host cells. Mononuclear phagocytes are actively inhibited from engulfing healthy cells via repulsive signals, which can be lost from the cell surface or attenuate their avidity during cell injury or apoptosis. Interestingly, recent work has shown that gene encoded variation in scavenger receptors can result in the abrogation or altered engagement of ‘don’t eat me’ signals, resulting in the recognition of non-self. Patterns associated with death also can be utilized by the innate immune system to acquire alloantigens or inhibit inflammatory responses by APCs.
Signal regulatory protein α (SIRPα)
Signal regulatory protein α (also known as SIRPα, CD172a, SHPS-1) is a transmembrane glycoprotein expressed mainly by myeloid cells, neurons, and stem cells. Although originally described as an inhibitor of growth factor signaling [156], SIRPα was later shown to prevent macrophage phagocytosis of healthy host cells [157]. Negative regulation of phagocytosis is accomplished by two cytoplasmic immunoreceptor tyrosine inhibitory motifs which, when phosphorylated in response to ligand engagement, trigger the recruitment of src homology phosphatases that act to block the remodeling actin cytoskeleton required for phagocytic activity [158, 159]. SIRPα binds to the broadly expressed integrin CD47. During apoptosis, CD47 changes its cell surface localization from a punctate to diffuse distribution, which in turn decreases its avidity for SIRPα, resulting in the attenuation of repulsive signals that prevent phagocytic clearance [160].
Early studies analyzing macrophage phagocytosis of red blood cells and peripheral blood mononuclear cells isolated from discordant vertebrate species suggested that phylogenetically encoded differences in SIRPα direct self-recognition [161–163]. Those findings were later reflected in positional genetics studies utilizing the non-obese diabetic (NOD)-severe combined deficiency (SCID) xenotransplantation model [164]. NOD.SCID mice are commonly used for human hematopoietic stem cell (HSC) studies due to the ability of human HSCs to self-renew and differentiate on this background. NOD SIRPα demonstrated a strong affinity for human CD47, while SIRPα from other strains were found to be poor binders of human CD47 and could not support HSC engraftment. Additionally, it was later observed that pig HSCs engineered to express human CD47 (huCD47) engrafted efficiently into NOD mice [165].
SIRPα also has been implicated recently in the control of the survival of solid organ xenotransplants. Baboon SIRPα is known to recognize huCD47. In a mixed chimerism model, pig skin grafted onto baboons was sharply prolonged by the administration of huCD47+ HSCs [166]. Also, huCD47 expressed on pig lung xenografts prolonged survival in baboons [167], which was later shown to be further extended by preconditioning with huCD47+ pig bone marrow transplants [168]. Perhaps the most surprising finding is that SIRPα may also play a role in alloimmune responses. Using a mouse bone marrow plug transplantation model and positional cloning techniques, Lakkis and colleagues reported that donor SIRPα amino acid polymorphisms can serve as markers of non-self [169]. Interestingly, most amino acid variability within SIRPα mouse locus was within the extracellular region and was particularly prevalent in CD47-binding immunoglobulin-like variable domain. The importance of this observation was underscored by greater binding of CD47 to allelic variants of SIRPα that promoted monocyte proliferation and monocyte-derived dendritic cell generation within bone marrow plug allografts. The authors hypothesized that innate allorecognition occurred when donor SIRPα had a greater affinity for CD47 when compared to recipient SIRPα. Although SIRPα polymorphisms also exist in humans [164], it remains to be determined if such differences control clinical transplantation outcomes.
CD200
CD200 (also known as OX-2) is a member of the immunoglobulin superfamily primarily expressed in many cell types, including antigen presenting cells. During apoptosis, CD200 expression is upregulated through p53- and caspase-dependent pathways and reduces tissue injury and inflammatory cytokine expression [170]. The immunomodulatory effects of CD200 are mediated through its interaction with CD200R, expressed mainly by myeloid-derived APCs and activated T cells [171, 172]. CD200-deficient mice are reported to have elevated steady-state APC activation and higher susceptibility to collagen-induced arthritis and experimental autoimmune encephalitis, suggesting a critical role for this pathway in immune homeostasis [173]. In the transplant setting, early work with CD200 Fc fusion protein was shown to increase mouse skin and kidney allograft survival, and attenuate the production of antibodies to sheep red blood cell erythrocytes, indicating that CD200:CD200R signaling regulated both allo- and xenogeneic responses [174]. The effects of blocking CD200 appear to be mediated by blocking Th1 cytokine production and inducing regulatory CD4+ T cell expansion by conventional allostimulatory dendritic cells [175, 176]. Recent work with overexpression of human CD200 porcine endothelial cells (PEC) has indicated its possible use in preventing xenograft rejection in humans [177]. Overexpression of CD200 in PECs suppressed inflammatory responses by human macrophages, including TNFα, IL-1β and IL-6 expression, and improved the survival of PEC xenografts in NOD.SCID mice.
CD36
CD36 is a widely expressed transmembrane glycoprotein scavenger receptor that serves many functions in lipid metabolism and signaling. It is a well-established mediator apoptotic cell clearance through helping recognition of oxidized phosphatidyl serine, a lipid marker of cellular senescence [178, 179]. CD36 on dendritic cells promotes cross-presentation of antigens from apoptotic cells to cytotoxic cells, suggesting that CD36 promotes alloreactivity by presenting antigen derived from phagocytized apoptotic graft cells [180]. However, recent evidence suggests CD36 expression on dendritic cells may also have regulatory effects. Work conducted by Hsieh and colleagues, using a mouse allogeneic bone marrow transplant model, demonstrated that CD36 facilitated the transfer of intact MHC:peptide complexes from apoptotic bodies released by thymic epithelial cells to CD8a+ dendritic cells, which in turn altered the thymic regulatory T cell repertoire [181]. They also observed decreases in CD8a+ dendritic cells and CD36 expression in the peripheral blood of human bone marrow recipients with graft-versus host disease.
TAMs
TAMs are a distinct group of tyrosine receptor kinases found on APCs that derive their name from three family members - Tyro3, Axl, and Mer. TAMs promote efferocytosis, a phagocytotic process that facilitates the removal of dying cells. TAMs interact with the TAM ligands growth arrest-specific gene 6 protein [182] and Protein S [183], that in turn recognize phosphatidyl serine [184]. TAM engagement promotes the clearance of dying cells through a poorly described signaling pathway that results in protein kinase C activation, which stimulates actin cytoskeleton remodeling necessary for phagocytosis [185]. TAMs also help resolve inflammation through complexing with the type I interferon receptor, which results in the attenuation of type I interferon and TLR-induced inflammatory cytokine production through driving the expression of suppressor of cytokine signaling 1 and 3 (SOCS1, SOCS3) [186]. Consistent with these observations is the profound dysregulation of immune responses and loss of self-tolerance in mice deficient in one or multiple TAMs [187, 188]. Infusion of apoptotic donor cells into murine models of islet [189] and heart [190] transplantation has suggested the importance of efferocytosis in regulating transplantation tolerance. These observations may also extend to the clinic. Extracorporeal photopheresis (ECP) is an FDA approved immunosuppressive therapy that utilizes ultraviolet A light–activated 8-methoxypsoralen-treated autologous leukocytes that are reinfused into a patient. ECP has been used to treat GVHD [191] and bronchiolitis obliterans syndrome [192], a form of chronic rejection in lung transplant recipients. The ECP treatment produces apoptotic cells, which are thought to drive some of its immunomodulatory effects [193]. Moreover, single infusions of autologous apoptotic mononuclear cells into patients with GVHD have been reported to inhibit dendritic cell maturation following challenge with LPS [194]. However, whether specific TAMs are involved in promoting allograft tolerance has only recently been investigated. Thorpe and colleagues demonstrated that the TAM Mer (MerTK) is required for mouse cardiac allograft tolerance driven by the infusion of donor apoptotic splenocytes [195].
Concluding Remarks
The transplantation of an organ into a host can be recognized by germline encoded PRRs that are stimulated by molecular patterns released or presented by injured tissue. Recent work has also supported the notion that PRRs may also recognize xeno- or allotypic differences, due to PRR polymorphisms that are present across phylogeny or within individuals of the same species. Resulting PRR engagement leads to the signaling pathways that drive the upregulation of inflammatory cytokines and antigen presentation molecules that promote adaptive immune responses against organs, ultimately leading to rejection. The selective targeting of PRRs could lead to the development of promising new therapies to promote immune tolerance.
Table 1.
PRR | Ligand | Organ | Injury |
---|---|---|---|
C1q | Fc region of IgM, IgG Annexin A2, A5 Membrane receptors on cells (e.g., calreticulin) | Heart | Protects against acute rejection by diminishing the humoral response [11] |
Skin | Protects against chronic rejection [49] [50] | ||
C-reactive protein (CRP) | C1q, phosphocholine, modified LDL, certain polycations, extracellular matrix proteins (e.g., fibronectin) | Kidney | Worsens IRI [62] [63] [64] |
Liver | Associated with worse IRI [65] | ||
Pentraxin-3 (PTX3) | C1q, growth factors (i.e., FGF2), extracellular matrix components (i.e. TSG-6), late apoptotic cells, pathogens, endothelial cells, leukocytes | Intestine | Worsens IRI [196] |
Heart | Cardioprotective role in IRI [70] [71] | ||
Kidney | Protective role in IRI [72, 73] | ||
Mannose-binding lectin (MBL) | Bacterial carbohydrate motifs, Fc region of bound immunoglobulin molecules | Heart | - Worsens IRI [89] [90] [91] - Accelerates chronic allograft loss post-cold ischemia [93] |
Kidney | Worsens IRI [197] [94] [95] [198] | ||
Gut | Worsens IRI [199] | ||
MBL-associated serine protease-2 (MASP2) | Enzyme binding to MBL | Kidney | Worsens DGF [102] |
Heart | Worsens IRI [101] | ||
Gut | Worsens IRI [101] | ||
Collectin-11 (CL-11) | L-fucose | Kidney | Worsens IRI [75] |
Surfactant protein A (SP-A) | Wide range of microorganisms and apoptotic cells via receptors (i.e., TLR4, C1qR, SIRPα) | Lung | - Reduces IRI in rats [82] [83] |
Abbreviations: FGF2: fibroblast growth factor-2; IRI: ischemia-reperfusion injury; LDL: lowdensity lipoprotein; SIRP-α: Signal regulatory protein α; TLR4: Toll-like receptor 4; TSG-6: TNF-stimulated gene 6 protein
Table 3.
Pattern Recognition Molecule | Ligand | Target Organ | Injury |
---|---|---|---|
SIRPα | CD47 (broadly expressed)s | Skin | - Pig skin grafted onto baboons sharply prolonged by the administration of pig huCD47+ HSCs (mixed chimerism model) [166]. - huCD47 expressed on pig lung xenografts prolonged survival in baboons [167], which could be further extended by preconditioning baboon recipients with pig bone marrow transplants bearing huCD47 [168]. |
Bone marrow | - Greater binding of CD47 to allelic variants of Sirp1a promoted monocyte proliferation and monocyte-derived dendritic cell generation within bone marrow plug allografts [169]. | ||
CD200 | CD200R (myeloid-derived APCs and activated T cells) |
Skin | - Treatment with CD200 Fc fusion protein increases mouse skin allograft survival [174]. |
Kidney | - Treatment with CD200 Fc fusion protein increases mouse kidney allograft survival [174]. | ||
Xenografts | - Overexpression of CD200 in porcine endothelial cells (PEC) suppressed inflammatory responses by human macrophages (i.e., TNFα, IL-1β and IL-6), and improved survival of PEC xenografts in NOD.SCID mice. [177]. | ||
CD36 | Oxidized phosphatidyl serine | Bone marrow | - CD36 facilitates allotolerance by the transfer of intact MHC:peptide complexes from apoptotic bodies released by thymic epithelial cells to CD8a+ dendritic cells, which in turn altered the thymic regulatory T cell repertoire [181]. |
MerTK | Protein S, Gas6 | Heart | - Facilitates tolerance driven by the infusion of donor apoptotic splenocytes [195]. |
Abbreviations: APC: antigen-presenting cells; MHC: major histocompatibility complex
Acknowledgements
This work was supported by the Barnes-Jewish Foundation, a Cystic Fibrosis Foundation research grant, and NIH grants, P01AI116501–01, R21AI119506, and 2RHL094601.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Smith NC, Rise ML, Christian SL, A Comparison of the Innate and Adaptive Immune Systems in Cartilaginous Fish, Ray-Finned Fish, and Lobe-Finned Fish, Front Immunol, 10 (2019) 2292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Zhou W, Farrar CA, Abe K, Pratt JR, Marsh JE, Wang Y, Stahl GL, Sacks SH, Predominant role for C5b-9 in renal ischemia/reperfusion injury, J Clin Invest, 105 (2000) 1363–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Lin T, Zhou W, Farrar CA, Hargreaves RE, Sheerin NS, Sacks SH, Deficiency of C4 from donor or recipient mouse fails to prevent renal allograft rejection, The American journal of pathology, 168 (2006) 1241–1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Diepenhorst GM, van Gulik TM, Hack CE, Complement-mediated ischemia-reperfusion injury: lessons learned from animal and clinical studies, Annals of surgery, 249 (2009) 889–899. [DOI] [PubMed] [Google Scholar]
- [5].Thurman JM, Ljubanovic D, Edelstein CL, Gilkeson GS, Holers VM, Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice, J Immunol, 170 (2003) 1517–1523. [DOI] [PubMed] [Google Scholar]
- [6].Casiraghi F, Azzollini N, Todeschini M, Fiori S, Cavinato RA, Cassis P, Solini S, Pezzuto F, Mister M, Thurman JM, Benigni A, Remuzzi G, Noris M, Complement Alternative Pathway Deficiency in Recipients Protects Kidney Allograft From Ischemia/Reperfusion Injury and Alloreactive T Cell Response, Am J Transplant, 17 (2017) 2312–2325. [DOI] [PubMed] [Google Scholar]
- [7].Bongoni AK, Kiermeir D, Jenni H, Wunsch A, Bahr A, Ayares D, Seebach JD, Wolf E, Klymiuk N, Constantinescu MA, Vogelin E, Rieben R, Activation of the lectin pathway of complement in pig-to-human xenotransplantation models, Transplantation, 96 (2013) 791–799. [DOI] [PubMed] [Google Scholar]
- [8].de Oliveira THC, Souza DG, Teixeira MM, Amaral FA, Tissue Dependent Role of PTX3 During Ischemia-Reperfusion Injury, Front Immunol, 10 (2019) 1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Agashe VV, Jankowska-Gan E, Keller M, Sullivan JA, Haynes LD, Kernien JF, Torrealba JR, Roenneburg D, Dart M, Colonna M, Wilkes DS, Burlingham WJ, Leukocyte-Associated Ig-like Receptor 1 Inhibits Th1 Responses but Is Required for Natural and Induced Monocyte-Dependent Th17 Responses, J Immunol, 201 (2018) 772–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Baruah P, Simpson E, Dumitriu IE, Derbyshire K, Coe D, Addey C, Dyson J, Chai J-G, Cook T, Scott D, Botto M, Mice lacking C1q or C3 show accelerated rejection of minor H disparate skin grafts and resistance to induction of tolerance, Eur J Immunol, 40 (2010) 1758–1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Csencsits K, Burrell BE, Lu G, Eichwald EJ, Stahl GL, Bishop DK, The classical complement pathway in transplantation: unanticipated protective effects of C1q and role in inductive antibody therapy, Am J Transplant, 8 (2008) 1622–1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Murata K, Fox-Talbot K, Qian Z, Takahashi K, Stahl GL, Baldwin WM 3rd, Wasowska BA, Synergistic deposition of C4d by complement-activating and non-activating antibodies in cardiac transplants, Am J Transplant, 7 (2007) 2605–2614. [DOI] [PubMed] [Google Scholar]
- [13].Munster JM, van der Bij W, Breukink MB, van der Steege G, Zuurman MW, Hepkema BG, Verschuuren EA, van Son WJ, Seelen MA, Association between donor MBL promoter haplotype and graft survival and the development of BOS after lung transplantation, Transplantation, 86 (2008) 1857–1863. [DOI] [PubMed] [Google Scholar]
- [14].Wu W, Liu C, Farrar CA, Ma L, Dong X, Sacks SH, Li K, Zhou W, Collectin-11 Promotes the Development of Renal Tubulointerstitial Fibrosis, Journal of the American Society of Nephrology : JASN, 29 (2018) 168–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Damman J, Daha MR, van Son WJ, Leuvenink HG, Ploeg RJ, Seelen MA, Crosstalk between complement and Toll-like receptor activation in relation to donor brain death and renal ischemia-reperfusion injury, Am J Transplant, 11 (2011) 660–669. [DOI] [PubMed] [Google Scholar]
- [16].Jane-Wit D, Manes TD, Yi T, Qin L, Clark P, Kirkiles-Smith NC, Abrahimi P, Devalliere J, Moeckel G, Kulkarni S, Tellides G, Pober JS, Alloantibody and complement promote T cell-mediated cardiac allograft vasculopathy through noncanonical nuclear factor-kappaB signaling in endothelial cells, Circulation, 128 (2013) 2504–2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Sheen JH, Strainic MG, Liu J, Zhang W, Yi Z, Medof ME, Heeger PS, TLR-Induced Murine Dendritic Cell (DC) Activation Requires DC-Intrinsic Complement, J Immunol, 199 (2017) 278–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Khan MA, Jiang X, Dhillon G, Beilke J, Holers VM, Atkinson C, Tomlinson S, Nicolls MR, CD4+ T cells and complement independently mediate graft ischemia in the rejection of mouse orthotopic tracheal transplants, Circulation research, 109 (2011) 1290–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Qin L, Li G, Kirkiles-Smith N, Clark P, Fang C, Wang Y, Yu ZX, Devore D, Tellides G, Pober JS, Jane-Wit D, Complement C5 Inhibition Reduces T Cell-Mediated Allograft Vasculopathy Caused by Both Alloantibody and Ischemia Reperfusion Injury in Humanized Mice, Am J Transplant, 16 (2016) 2865–2876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Demir AVD,M, Chun N, Fribourg M, Cravedi P, Llaudo I, Woodruff TM, Yadav P, Lira SA, Medof ME, Heeger PS, T Cell Expression of C5a Receptor 2 Augments Murine Regulatory T Cell (TREG) Generation and TREG-Dependent Cardiac Allograft Survival, J Immunol, 200 (2018) 2186–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Llaudo I, Fribourg M, Medof ME, Conde P, Ochando J, Heeger PS, C5aR1 regulates migration of suppressive myeloid cells required for costimulatory blockade-induced murine allograft survival, Am J Transplant, 19 (2019) 633–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Vieyra M, Leisman S, Raedler H, Kwan WH, Yang M, Strainic MG, Medof ME, Heeger PS, Complement regulates CD4 T-cell help to CD8 T cells required for murine allograft rejection, The American journal of pathology, 179 (2011) 766–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Verghese DA, Chun N, Paz K, Fribourg M, Woodruff TM, Flynn R, Hu Y, Xiong H, Zhang W, Yi Z, Du J, Blazar BR, Heeger PS, C5aR1 regulates T follicular helper differentiation and chronic graftversus-host disease bronchiolitis obliterans, JCI insight, 3 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Mathern DR, Heeger KHJ,PS, Absence of recipient C3aR1 signaling limits expansion and differentiation of alloreactive CD8(+) T cell immunity and prolongs murine cardiac allograft survival, Am J Transplant, 19 (2019) 1628–1640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Fryer JP, Leventhal JR, Pao W, Stadler C, Jones M, Walsh T, Zhong R, Zhang Z, Wang H, Goodman DJ, Kurek M, d’Apice AJ, Blondin B, Ivancic D, Buckingham F, Kaufman D, Abecassis M, Stuart F, Anderson BE, Synthetic peptides which inhibit the interaction between C1q and immunoglobulin and prolong xenograft survival, Transplantation, 70 (2000) 828–836. [DOI] [PubMed] [Google Scholar]
- [26].van Zanden JE, Jager NM, Daha MR, Erasmus ME, Leuvenink HGD, Seelen MA, Complement Therapeutics in the Multi-Organ Donor: Do or Don’t?, Front Immunol, 10 (2019) 329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Poppelaars F, Jager NM, Kotimaa J, Leuvenink HGD, Daha MR, van Kooten C, Seelen MA, Damman J, C1-Inhibitor Treatment Decreases Renal Injury in an Established Brain-Dead Rat Model, Transplantation, 102 (2018) 79–87. [DOI] [PubMed] [Google Scholar]
- [28].Khan MA, Maasch C, Vater A, Klussmann S, Morser J, Leung LL, Atkinson C, Tomlinson S, Heeger PS, Nicolls MR, Targeting complement component 5a promotes vascular integrity and limits airway remodeling, Proceedings of the National Academy of Sciences of the United States of America, 110 (2013) 6061–6066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Zhu P, Bailey SR, Lei B, Paulos CM, Atkinson C, Tomlinson S, Targeted Complement Inhibition Protects Vascularized Composite Allografts From Acute Graft Injury and Prolongs Graft Survival When Combined With Subtherapeutic Cyclosporine A Therapy, Transplantation, 101 (2017) e75–e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Keshavjee S, Davis RD, Zamora MR, de Perrot M, Patterson GA, A randomized, placebo-controlled trial of complement inhibition in ischemia-reperfusion injury after lung transplantation in human beings, J Thorac Cardiovasc Surg, 129 (2005) 423–428. [DOI] [PubMed] [Google Scholar]
- [31].Kulkarni HS, Liszewski MK, Brody SL, Atkinson JP, The complement system in the airway epithelium: An overlooked host defense mechanism and therapeutic target?, The Journal of allergy and clinical immunology, 141 (2018) 1582–1586.e1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Sheerin NS, Risley P, Abe K, Tang Z, Wong W, Lin T, Sacks SH, Synthesis of complement protein C3 in the kidney is an important mediator of local tissue injury, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 22 (2008) 1065–1072. [DOI] [PubMed] [Google Scholar]
- [33].Fuquay R, Renner B, Kulik L, McCullough JW, Amura C, Strassheim D, Pelanda R, Torres R, Thurman JM, Renal ischemia-reperfusion injury amplifies the humoral immune response, Journal of the American Society of Nephrology : JASN, 24 (2013) 1063–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Strainic MG, Shevach EM, An F, Lin F, Medof ME, Absence of signaling into CD4(+) cells via C3aR and C5aR enables autoinductive TGF-beta1 signaling and induction of Foxp3(+) regulatory T cells, Nat Immunol, 14 (2013) 162–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Cravedi P, Leventhal J, Lakhani P, Ward SC, Donovan MJ, Heeger PS, Immune cell-derived C3a and C5a costimulate human T cell alloimmunity, Am J Transplant, 13 (2013) 2530–2539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Kwan WH, van der Touw W, Paz-Artal E, Li MO, Heeger PS, Signaling through C5a receptor and C3a receptor diminishes function of murine natural regulatory T cells, The Journal of experimental medicine, 210 (2013) 257–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Triantafilou M, Hughes TR, Morgan BP, Triantafilou K, Complementing the inflammasome, Immunology, 147 (2016) 152–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Jane-Wit D, Manes TD, Yi T, Qin L, Clark P, Kirkiles-Smith NC, Abrahimi P, Devalliere J, Moeckel G, Kulkarni S, Tellides G, Pober JS, Alloantibody and complement promote T cell-mediated cardiac allograft vasculopathy through noncanonical nuclear factor-κB signaling in endothelial cells, Circulation, 128 (2013) 2504–2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Thurman JM, Ljubanović D, Royer PA, Kraus DM, Molina H, Barry NP, Proctor G, Levi M, Holers VM, Altered renal tubular expression of the complement inhibitor Crry permits complement activation after ischemia/reperfusion, The Journal of clinical investigation, 116 (2006) 357–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Gal P, Dobo J, Zavodszky P, Sim RB, Early complement proteases: C1r, C1s and MASPs. A structural insight into activation and functions, Molecular immunology, 46 (2009) 2745–2752. [DOI] [PubMed] [Google Scholar]
- [41].Thielens NM, Tedesco F, Bohlson SS, Gaboriaud C, Tenner AJ, C1q: A fresh look upon an old molecule, Molecular immunology, 89 (2017) 73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Martin M, Leffler J, Blom AM, Annexin A2 and A5 serve as new ligands for C1q on apoptotic cells, The Journal of biological chemistry, 287 (2012) 33733–33744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Scott D, Botto M, The paradoxical roles of C1q and C3 in autoimmunity, Immunobiology, 221 (2016) 719–725. [DOI] [PubMed] [Google Scholar]
- [44].Cortes-Hernandez J, Fossati-Jimack L, Petry F, Loos M, Izui S, Walport MJ, Cook HT, Botto M, Restoration of C1q levels by bone marrow transplantation attenuates autoimmune disease associated with C1q deficiency in mice, Eur J Immunol, 34 (2004) 3713–3722. [DOI] [PubMed] [Google Scholar]
- [45].Petry F, Botto M, Holtappels R, Walport MJ, Loos M, Reconstitution of the complement function in C1q-deficient (C1qa−/−) mice with wild-type bone marrow cells, J Immunol, 167 (2001) 4033–4037. [DOI] [PubMed] [Google Scholar]
- [46].Ling GS, Crawford G, Buang N, Bartok I, Tian K, Thielens NM, Bally I, Harker JA, Ashton-Rickardt PG, Rutschmann S, Strid J, Botto M, C1q restrains autoimmunity and viral infection by regulating CD8(+) T cell metabolism, Science (New York, N.Y.), 360 (2018) 558–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Clarke EV, Weist BM, Walsh CM, Tenner AJ, Complement protein C1q bound to apoptotic cells suppresses human macrophage and dendritic cell-mediated Th17 and Th1 T cell subset proliferation, Journal of leukocyte biology, 97 (2015) 147–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Benoit ME, Clarke EV, Morgado P, Fraser DA, Tenner AJ, Complement protein C1q directs macrophage polarization and limits inflammasome activity during the uptake of apoptotic cells, J Immunol, 188 (2012) 5682–5693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Baruah P, Simpson E, Dumitriu IE, Derbyshire K, Coe D, Addey C, Dyson J, Chai JG, Cook T, Scott D, Botto M, Mice lacking C1q or C3 show accelerated rejection of minor H disparate skin grafts and resistance to induction of tolerance, Eur J Immunol, 40 (2010) 1758–1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Fossati-Jimack L, Ling GS, Baudino L, Szajna M, Manivannan K, Zhao JC, Midgley R, Chai JG, Simpson E, Botto M, Scott D, Intranasal peptide-induced tolerance and linked suppression: consequences of complement deficiency, Immunology, 144 (2015) 149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Naito AT, Sumida T, Nomura S, Liu ML, Higo T, Nakagawa A, Okada K, Sakai T, Hashimoto A, Hara Y, Shimizu I, Zhu W, Toko H, Katada A, Akazawa H, Oka T, Lee JK, Minamino T, Nagai T, Walsh K, Kikuchi A, Matsumoto M, Botto M, Shiojima I, Komuro I, Complement C1q activates canonical Wnt signaling and promotes aging-related phenotypes, Cell, 149 (2012) 1298–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Ma YJ, Lee BL, Garred P, An overview of the synergy and crosstalk between pentraxins and collectins/ficolins: their functional relevance in complement activation, Experimental & molecular medicine, 49 (2017) e320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Thomsen T, Schlosser A, Holmskov U, Sorensen GL, Ficolins and FIBCD1: soluble and membrane bound pattern recognition molecules with acetyl group selectivity, Molecular immunology, 48 (2011) 369–381. [DOI] [PubMed] [Google Scholar]
- [54].Eikmans M, de Canck I, van der Pol P, Baan CC, Haasnoot GW, Mallat MJ, Vergunst M, de Meester E, Roodnat JI, Anholts JD, van Thielen M, Doxiadis II, de Fijter JW, van der Linden PJ, van Beelen E, van Kooten C, Kal-van Gestel JA, Peeters AM, Weimar W, Roelen DL, Rossau R, Claas FH, The functional polymorphism Ala258Ser in the innate receptor gene ficolin-2 in the donor predicts improved renal transplant outcome, Transplantation, 94 (2012) 478–485. [DOI] [PubMed] [Google Scholar]
- [55].Smedbraten YV, Sagedal S, Mjoen G, Hartmann A, Fagerland MW, Rollag H, Mollnes TE, Thiel S, High ficolin-3 level at the time of transplantation is an independent risk factor for graft loss in kidney transplant recipients, Transplantation, 99 (2015) 791–796. [DOI] [PubMed] [Google Scholar]
- [56].Daigo K, Inforzato A, Barajon I, Garlanda C, Bottazzi B, Meri S, Mantovani A, Pentraxins in the activation and regulation of innate immunity, Immunological reviews, 274 (2016) 202–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Mantovani A, Garlanda C, Doni A, Bottazzi B, Pentraxins in innate immunity: from C-reactive protein to the long pentraxin PTX3, Journal of clinical immunology, 28 (2008) 1–13. [DOI] [PubMed] [Google Scholar]
- [58].Doni A, Stravalaci M, Inforzato A, Magrini E, Mantovani A, Garlanda C, Bottazzi B, The Long Pentraxin PTX3 as a Link Between Innate Immunity, Tissue Remodeling, and Cancer, Front Immunol, 10 (2019) 712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Du Clos TW, Mold C, Pentraxins (CRP, SAP) in the process of complement activation and clearance of apoptotic bodies through Fcgamma receptors, Current opinion in organ transplantation, 16 (2011) 15–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Xing D, Hage FG, Chen YF, McCrory MA, Feng W, Skibinski GA, Majid-Hassan E, Oparil S, Szalai AJ, Exaggerated neointima formation in human C-reactive protein transgenic mice is IgG Fc receptor type I (Fc gamma RI)-dependent, The American journal of pathology, 172 (2008) 22–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Hage FG, Oparil S, Xing D, Chen YF, McCrory MA, Szalai AJ, C-reactive protein-mediated vascular injury requires complement, Arteriosclerosis, thrombosis, and vascular biology, 30 (2010) 1189–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Pegues MA, McCrory MA, Zarjou A, Szalai AJ, C-reactive protein exacerbates renal ischemia-reperfusion injury, American journal of physiology. Renal physiology, 304 (2013) F1358–1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Bian A, Shi M, Flores B, Gillings N, Li P, Yan SX, Levine B, Xing C, Hu MC, Downregulation of autophagy is associated with severe ischemia-reperfusion-induced acute kidney injury in overexpressing C-reactive protein mice, PLoS One, 12 (2017) e0181848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Thiele JR, Zeller J, Kiefer J, Braig D, Kreuzaler S, Lenz Y, Potempa LA, Grahammer F, Huber TB, Huber-Lang M, Bannasch H, Stark GB, Peter K, Eisenhardt SU, A Conformational Change in C-Reactive Protein Enhances Leukocyte Recruitment and Reactive Oxygen Species Generation in Ischemia/Reperfusion Injury, Front Immunol, 9 (2018) 675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Diepenhorst GM, de Graaf W, Niessen HW, van Vliet AK, Hack CE, van Gulik TM, Immunoglobulin M, C-reactive protein and complement activation in rat hepatic ischemia-reperfusion injury, European surgical research. Europaische chirurgische Forschung. Recherches chirurgicales europeennes, 52 (2014) 50–62. [DOI] [PubMed] [Google Scholar]
- [66].Doni A, Garlanda C, Bottazzi B, Meri S, Garred P, Mantovani A, Interactions of the humoral pattern recognition molecule PTX3 with the complement system, Immunobiology, 217 (2012) 1122–1128. [DOI] [PubMed] [Google Scholar]
- [67].Baruah P, Dumitriu IE, Peri G, Russo V, Mantovani A, Manfredi AA, Rovere-Querini P, The tissue pentraxin PTX3 limits C1q-mediated complement activation and phagocytosis of apoptotic cells by dendritic cells, Journal of leukocyte biology, 80 (2006) 87–95. [DOI] [PubMed] [Google Scholar]
- [68].Lu J, Mold C, Du Clos TW, Sun PD, Pentraxins and Fc Receptor-Mediated Immune Responses, Front Immunol, 9 (2018) 2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Salio M, Chimenti S, De Angelis N, Molla F, Maina V, Nebuloni M, Pasqualini F, Latini R, Garlanda C, Mantovani A, Cardioprotective function of the long pentraxin PTX3 in acute myocardial infarction, Circulation, 117 (2008) 1055–1064. [DOI] [PubMed] [Google Scholar]
- [70].Zhu H, Cui D, Liu K, Wang L, Huang L, Li J, Long pentraxin PTX3 attenuates ischemia reperfusion injury in a cardiac transplantation model, Transplant international : official journal of the European Society for Organ Transplantation, 27 (2014) 87–95. [DOI] [PubMed] [Google Scholar]
- [71].Shimizu T, Suzuki S, Sato A, Nakamura Y, Ikeda K, Saitoh S, Misaka S, Shishido T, Kubota I, Takeishi Y, Cardio-protective effects of pentraxin 3 produced from bone marrow-derived cells against ischemia/reperfusion injury, Journal of molecular and cellular cardiology, 89 (2015) 306–313. [DOI] [PubMed] [Google Scholar]
- [72].Xiao Y, Yang N, Zhang Q, Wang Y, Yang S, Liu Z, Pentraxin 3 inhibits acute renal injury-induced interstitial fibrosis through suppression of IL-6/Stat3 pathway, Inflammation, 37 (2014) 1895–1901. [DOI] [PubMed] [Google Scholar]
- [73].Lech M, Rommele C, Grobmayr R, Eka Susanti H, Kulkarni OP, Wang S, Grone HJ, Uhl B, Reichel C, Krombach F, Garlanda C, Mantovani A, Anders HJ, Endogenous and exogenous pentraxin-3 limits postischemic acute and chronic kidney injury, Kidney Int, 83 (2013) 647–661. [DOI] [PubMed] [Google Scholar]
- [74].Howard M, Farrar CA, Sacks SH, Structural and functional diversity of collectins and ficolins and their relationship to disease, Seminars in immunopathology, 40 (2018) 75–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Farrar CA, Tran D, Li K, Wu W, Peng Q, Schwaeble W, Zhou W, Sacks SH, Collectin-11 detects stress-induced L-fucose pattern to trigger renal epithelial injury, J Clin Invest, 126 (2016) 1911–1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Watford WT, Wright JR, Hester CG, Jiang H, Frank MM, Surfactant protein A regulates complement activation, J Immunol, 167 (2001) 6593–6600. [DOI] [PubMed] [Google Scholar]
- [77].Yang S, Milla C, Panoskaltsis-Mortari A, Ingbar DH, Blazar BR, Haddad IY, Human surfactant protein a suppresses T cell-dependent inflammation and attenuates the manifestations of idiopathic pneumonia syndrome in mice, American journal of respiratory cell and molecular biology, 24 (2001) 527–536. [DOI] [PubMed] [Google Scholar]
- [78].Jiaravuthisan P, Maeda A, Takakura C, Wang HT, Sakai R, Shabri AM, Lo PC, Matsuura R, Kodama T, Eguchi H, Okuyama H, Miyagawa S, A membrane-type surfactant protein D (SP-D) suppresses macrophage-mediated cytotoxicity in swine endothelial cells, Transpl Immunol, 47 (2018) 44–48. [DOI] [PubMed] [Google Scholar]
- [79].D’Ovidio F, Kaneda H, Chaparro C, Mura M, Lederer D, Di Angelo S, Takahashi H, Gutierrez C, Hutcheon M, Singer LG, Waddell TK, Floros J, Liu M, Keshavjee S, Pilot study exploring lung allograft surfactant protein A (SP-A) expression in association with lung transplant outcome, Am J Transplant, 13 (2013) 2722–2729. [DOI] [PubMed] [Google Scholar]
- [80].D’Ovidio F, Floros J, Aramini B, Lederer D, DiAngelo SL, Arcasoy S, Sonett JR, Robbins H, Shah L, Costa J, Urso A, Donor Surfactant Protein A2 Polymorphism and Lung Transplant Survival, The European respiratory journal, (2019). [DOI] [PubMed] [Google Scholar]
- [81].Belhaj A, Boven C, Dewachter L, Ruiz Patino M, Sokolow Y, Rondelet B, Influence of Donor Lung Surfactant-A and -B Protein Expression on the Development of Primary Graft Dysfunction After Lung Transplantation: A Pilot Study, Annals of transplantation, 22 (2017) 361–369. [DOI] [PubMed] [Google Scholar]
- [82].Erasmus ME, Hofstede GJ, Petersen AH, Haagsman HP, Oetomo SB, Prop J, Effects of early surfactant treatment persisting for one week after lung transplantation in rats, Am J Respir Crit Care Med, 156 (1997) 567–572. [DOI] [PubMed] [Google Scholar]
- [83].Erasmus ME, Hofstede GJ, Petersen AH, Batenburg JJ, Haagsman HP, Oetomo SB, Prop J, SP-A-enriched surfactant for treatment of rat lung transplants with SP-A deficiency after storage and reperfusion, Transplantation, 73 (2002) 348–352. [DOI] [PubMed] [Google Scholar]
- [84].Garred P, Genster N, Pilely K, Bayarri-Olmos R, Rosbjerg A, Ma YJ, Skjoedt MO, A journey through the lectin pathway of complement-MBL and beyond, Immunological reviews, 274 (2016) 74–97. [DOI] [PubMed] [Google Scholar]
- [85].Tenner AJ, C1q receptors: regulating specific functions of phagocytic cells, Immunobiology, 199 (1998) 250–264. [DOI] [PubMed] [Google Scholar]
- [86].Kilpatrick DC, Stewart K, Allan EK, McLintock LA, Holyoake TL, Turner ML, Successful haemopoietic stem cell transplantation does not correct mannan-binding lectin deficiency, Bone marrow transplantation, 35 (2005) 179–181. [DOI] [PubMed] [Google Scholar]
- [87].Ibernon M, Moreso F, Seron D, Innate immunity in renal transplantation: the role of mannose-binding lectin, Transplantation reviews (Orlando, Fla.), 28 (2014) 21–25. [DOI] [PubMed] [Google Scholar]
- [88].Berger SP, Daha MR, Emerging role of the mannose-binding lectin-dependent pathway of complement activation in clinical organ transplantation, Current opinion in organ transplantation, 16 (2011) 28–33. [DOI] [PubMed] [Google Scholar]
- [89].Walsh MC, Bourcier T, Takahashi K, Shi L, Busche MN, Rother RP, Solomon SD, Ezekowitz RA, Stahl GL, Mannose-binding lectin is a regulator of inflammation that accompanies myocardial ischemia and reperfusion injury, J Immunol, 175 (2005) 541–546. [DOI] [PubMed] [Google Scholar]
- [90].Busche MN, Pavlov V, Takahashi K, Stahl GL, Myocardial ischemia and reperfusion injury is dependent on both IgM and mannose-binding lectin, American journal of physiology. Heart and circulatory physiology, 297 (2009) H1853–1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].La Bonte LR, Dokken B, Davis-Gorman G, Stahl GL, McDonagh PF, The mannose-binding lectin pathway is a significant contributor to reperfusion injury in the type 2 diabetic heart, Diabetes & vascular disease research, 6 (2009) 172–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Xie CB, Qin L, Li G, Fang C, Kirkiles-Smith NC, Tellides G, Pober JS, Jane-Wit D, Complement Membrane Attack Complexes Assemble NLRP3 Inflammasomes Triggering IL-1 Activation of IFN-gamma-Primed Human Endothelium, Circulation research, 124 (2019) 1747–1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Chun N, Fairchild RL, Li Y, Liu J, Zhang M, Baldwin WM 3rd, Heeger PS, Complement Dependence of Murine Costimulatory Blockade-Resistant Cellular Cardiac Allograft Rejection, Am J Transplant, 17 (2017) 2810–2819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Castellano G, Melchiorre R, Loverre A, Ditonno P, Montinaro V, Rossini M, Divella C, Battaglia M, Lucarelli G, Annunziata G, Palazzo S, Selvaggi FP, Staffieri F, Crovace A, Daha MR, Mannesse M, van Wetering S, Paolo Schena F, Grandaliano G, Therapeutic targeting of classical and lectin pathways of complement protects from ischemia-reperfusion-induced renal damage, The American journal of pathology, 176 (2010) 1648–1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].van der Pol P, Schlagwein N, van Gijlswijk DJ, Berger SP, Roos A, Bajema IM, de Boer HC, de Fijter JW, Stahl GL, Daha MR, van Kooten C, Mannan-binding lectin mediates renal ischemia/reperfusion injury independent of complement activation, Am J Transplant, 12 (2012) 877–887. [DOI] [PubMed] [Google Scholar]
- [96].Davis AE 3rd, Lu F, Mejia P, C1 inhibitor, a multi-functional serine protease inhibitor, Thrombosis and haemostasis, 104 (2010) 886–893. [DOI] [PubMed] [Google Scholar]
- [97].Berger M, Baldwin WM 3rd, Jordan SC, Potential Roles for C1 Inhibitor in Transplantation, Transplantation, 100 (2016) 1415–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Panagiotou A, Trendelenburg M, Osthoff M, The Lectin Pathway of Complement in Myocardial Ischemia/Reperfusion Injury-Review of Its Significance and the Potential Impact of Therapeutic Interference by C1 Esterase Inhibitor, Front Immunol, 9 (2018) 1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Heja D, Kocsis A, Dobo J, Szilagyi K, Szasz R, Zavodszky P, Pal G, Gal P, Revised mechanism of complement lectin-pathway activation revealing the role of serine protease MASP-1 as the exclusive activator of MASP-2, Proceedings of the National Academy of Sciences of the United States of America, 109 (2012) 10498–10503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Ambrus G, Gal P, Kojima M, Szilagyi K, Balczer J, Antal J, Graf L, Laich A, Moffatt BE, Schwaeble W, Sim RB, Zavodszky P, Natural substrates and inhibitors of mannan-binding lectin-associated serine protease-1 and −2: a study on recombinant catalytic fragments, J Immunol, 170 (2003) 1374–1382. [DOI] [PubMed] [Google Scholar]
- [101].Schwaeble WJ, Lynch NJ, Clark JE, Marber M, Samani NJ, Ali YM, Dudler T, Parent B, Lhotta K, Wallis R, Farrar CA, Sacks S, Lee H, Zhang M, Iwaki D, Takahashi M, Fujita T, Tedford CE, Stover CM, Targeting of mannan-binding lectin-associated serine protease-2 confers protection from myocardial and gastrointestinal ischemia/reperfusion injury, Proceedings of the National Academy of Sciences of the United States of America, 108 (2011) 7523–7528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Asgari E, Farrar CA, Lynch N, Ali YM, Roscher S, Stover C, Zhou W, Schwaeble WJ, Sacks SH, Mannan-binding lectin-associated serine protease 2 is critical for the development of renal ischemia reperfusion injury and mediates tissue injury in the absence of complement C4, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 28 (2014) 3996–4003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].O’Neill LA, Golenbock D, Bowie AG, The history of Toll-like receptors - redefining innate immunity, Nat Rev Immunol, 13 (2013) 453–460. [DOI] [PubMed] [Google Scholar]
- [104].Kabelitz D, Expression and function of Toll-like receptors in T lymphocytes, Curr Opin Immunol, 19 (2007) 39–45. [DOI] [PubMed] [Google Scholar]
- [105].Janeway CA Jr., Approaching the asymptote? Evolution and revolution in immunology, Cold Spring Harbor symposia on quantitative biology, 54 Pt 1 (1989) 1–13. [DOI] [PubMed] [Google Scholar]
- [106].Medzhitov R, Preston-Hurlburt P, Janeway CA Jr., A human homologue of the Drosophila Toll protein signals activation of adaptive immunity, Nature, 388 (1997) 394–397. [DOI] [PubMed] [Google Scholar]
- [107].Matzinger P, The danger model: a renewed sense of self, Science (New York, N.Y.), 296 (2002) 301–305. [DOI] [PubMed] [Google Scholar]
- [108].Kawasaki T, Kawai T, Toll-like receptor signaling pathways, Front Immunol, 5 (2014) 461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Bell E, TLR4 signalling, Nature Reviews Immunology, 8 (2008) 241–241. [Google Scholar]
- [110].Goldstein DR, Tesar BM, Akira S, Lakkis FG, Critical role of the Toll-like receptor signal adaptor protein MyD88 in acute allograft rejection, J Clin Invest, 111 (2003) 1571–1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].McKay D, Shigeoka A, Rubinstein M, Surh C, Sprent J, Simultaneous deletion of MyD88 and Trif delays major histocompatibility and minor antigen mismatch allograft rejection, Eur J Immunol, 36 (2006) 1994–2002. [DOI] [PubMed] [Google Scholar]
- [112].Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, Tracey KJ, Chiorazzi N, High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization, J Immunol, 173 (2004) 307–313. [DOI] [PubMed] [Google Scholar]
- [113].McNulty S, Colaco CA, Blandford LE, Bailey CR, Baschieri S, Todryk S, Heat-shock proteins as dendritic cell-targeting vaccines--getting warmer, Immunology, 139 (2013) 407–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Johnson LA, Banerji S, Lawrance W, Gileadi U, Prota G, Holder KA, Roshorm YM, Hanke T, Cerundolo V, Gale NW, Jackson DG, Dendritic cells enter lymph vessels by hyaluronan-mediated docking to the endothelial receptor LYVE-1, Nat Immunol, 18 (2017) 762–770. [DOI] [PubMed] [Google Scholar]
- [115].Braza F, Brouard S, Chadban S, Goldstein DR, Role of TLRs and DAMPs in allograft inflammation and transplant outcomes, Nat Rev Nephrol, 12 (2016) 281–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Chong AJ, Shimamoto A, Hampton CR, Takayama H, Spring DJ, Rothnie CL, Yada M, Pohlman TH, Verrier ED, Toll-like receptor 4 mediates ischemia/reperfusion injury of the heart, J Thorac Cardiovasc Surg, 128 (2004) 170–179. [DOI] [PubMed] [Google Scholar]
- [117].Sakata Y, Dong JW, Vallejo JG, Huang CH, Baker JS, Tracey KJ, Tacheuchi O, Akira S, Mann DL, Toll-like receptor 2 modulates left ventricular function following ischemia-reperfusion injury, American journal of physiology. Heart and circulatory physiology, 292 (2007) H503–509. [DOI] [PubMed] [Google Scholar]
- [118].Zhai Y, Shen XD, O’Connell R, Gao F, Lassman C, Busuttil RW, Cheng G, Kupiec-Weglinski JW, Cutting edge: TLR4 activation mediates liver ischemia/reperfusion inflammatory response via IFN regulatory factor 3-dependent MyD88-independent pathway, J Immunol, 173 (2004) 7115–7119. [DOI] [PubMed] [Google Scholar]
- [119].Phelan P, Merry HE, Hwang B, Mulligan MS, Differential toll-like receptor activation in lung ischemia reperfusion injury, J Thorac Cardiovasc Surg, 149 (2015) 1653–1661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Shimamoto A, Pohlman TH, Shomura S, Tarukawa T, Takao M, Shimpo H, Toll-like receptor 4 mediates lung ischemia-reperfusion injury, The Annals of thoracic surgery, 82 (2006) 2017–2023. [DOI] [PubMed] [Google Scholar]
- [121].Wu H, Chen G, Wyburn KR, Yin J, Bertolino P, Eris JM, Alexander SI, Sharland AF, Chadban SJ, TLR4 activation mediates kidney ischemia/reperfusion injury, J Clin Invest, 117 (2007) 2847–2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Tesar BM, Jiang D, Liang J, Palmer SM, Noble PW, Goldstein DR, The role of hyaluronan degradation products as innate alloimmune agonists, Am J Transplant, 6 (2006) 2622–2635. [DOI] [PubMed] [Google Scholar]
- [123].Thornley TB, Brehm MA, Markees TG, Shultz LD, Mordes JP, Welsh RM, Rossini AA, Greiner DL, TLR agonists abrogate costimulation blockade-induced prolongation of skin allografts, J Immunol, 176 (2006) 1561–1570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Pasare C, Medzhitov R, Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells, Science (New York, N.Y.), 299 (2003) 1033–1036. [DOI] [PubMed] [Google Scholar]
- [125].Chen L, Ahmed E, Wang T, Wang Y, Ochando J, Chong AS, Alegre ML, TLR signals promote IL-6/IL-17-dependent transplant rejection, J Immunol, 182 (2009) 6217–6225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Porrett PM, Yuan X, LaRosa DF, Walsh PT, Yang J, Gao W, Li P, Zhang J, Ansari JM, Hancock WW, Sayegh MH, Koulmanda M, Strom TB, Turka LA, Mechanisms underlying blockade of allograft acceptance by TLR ligands, J Immunol, 181 (2008) 1692–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Bhat N, Fitzgerald KA, Recognition of cytosolic DNA by cGAS and other STING-dependent sensors, Eur J Immunol, 44 (2014) 634–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Zhang N, Kruger B, Lal G, Luan Y, Yadav A, Zang W, Grimm M, Waaga-Gasser AM, Murphy B, Bromberg JS, Schroppel B, Inhibition of TLR4 signaling prolongs Treg-dependent murine islet allograft survival, Immunol Lett, 127 (2010) 119–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Hsieh JL, Shen PC, Wu PT, Jou IM, Wu CL, Shiau AL, Wang CR, Chong HE, Chuang SH, Peng JS, Chen SY, Knockdown of toll-like receptor 4 signaling pathways ameliorate bone graft rejection in a mouse model of allograft transplantation, Sci Rep, 7 (2017) 46050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Sosa RA, Rossetti M, Naini BV, Groysberg VM, Kaldas FM, Busuttil RW, Chang YL, Gjertson DW, Kupiec-Weglinski JW, Reed EF, Pattern Recognition Receptor-reactivity Screening of Liver Transplant Patients: Potential for Personalized and Precise Organ Matching to Reduce Risks of Ischemiareperfusion Injury, Annals of surgery, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Hosseinzadeh M, Ahmadpoor P, Yekaninejad MS, Pourrezagholi F, Foroughi F, Ghorbanpour M, Barabadi M, Shahbaz SK, Solgi G, Amirzargar A, Expression patterns of Toll like receptor (TLR)-2, TLR-4 and myeloid differentiation primary response gene 88 (MYD88) in renal transplant patients developing allograft dysfunction; a cohort study, Transpl Immunol, 48 (2018) 26–31. [DOI] [PubMed] [Google Scholar]
- [132].Deng JF, Geng L, Qian YG, Li H, Wang Y, Xie HY, Feng XW, Zheng SS, The role of toll-like receptors 2 and 4 in acute allograft rejection after liver transplantation, Transplant Proc, 39 (2007) 3222–3224. [DOI] [PubMed] [Google Scholar]
- [133].Testro AG, Visvanathan K, Skinner N, Markovska V, Crowley P, Angus PW, Gow PJ, Acute allograft rejection in human liver transplant recipients is associated with signaling through toll-like receptor 4, J Gastroenterol Hepatol, 26 (2011) 155–163. [DOI] [PubMed] [Google Scholar]
- [134].Palmer SM, Burch LH, Davis RD, Herczyk WF, Howell DN, Reinsmoen NL, Schwartz DA, The role of innate immunity in acute allograft rejection after lung transplantation, Am J Respir Crit Care Med, 168 (2003) 628–632. [DOI] [PubMed] [Google Scholar]
- [135].Jia YP, Wang K, Zhang ZJ, Tong YN, Han D, Hu CY, Li Q, Xiang Y, Mao XH, Tang B, TLR2/TLR4 activation induces Tregs and suppresses intestinal inflammation caused by Fusobacterium nucleatum in vivo, PLoS One, 12 (2017) e0186179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Aumeunier A, Grela F, Ramadan A, Pham Van L, Bardel E, Gomez Alcala A, Jeannin P, Akira S, Bach JF, Thieblemont N, Systemic Toll-like receptor stimulation suppresses experimental allergic asthma and autoimmune diabetes in NOD mice, PLoS One, 5 (2010) e11484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Ray A, Chakraborty K, Ray P, Immunosuppressive MDSCs induced by TLR signaling during infection and role in resolution of inflammation, Front Cell Infect Microbiol, 3 (2013) 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Zhang W, Li J, Qi G, Tu G, Yang C, Xu M, Myeloid-derived suppressor cells in transplantation: the dawn of cell therapy, Journal of translational medicine, 16 (2018) 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Ochando J, Conde P, Utrero-Rico A, Paz-Artal E, Tolerogenic Role of Myeloid Suppressor Cells in Organ Transplantation, Front Immunol, 10 (2019) 374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Alegre ML, Leemans J, Le Moine A, Florquin S, De Wilde V, Chong A, Goldman M, The multiple facets of toll-like receptors in transplantation biology, Transplantation, 86 (2008) 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Jiang D, Liang J, Fan J, Yu S, Chen S, Luo Y, Prestwich GD, Mascarenhas MM, Garg HG, Quinn DA, Homer RJ, Goldstein DR, Bucala R, Lee PJ, Medzhitov R, Noble PW, Regulation of lung injury and repair by Toll-like receptors and hyaluronan, Nature Medicine, 11 (2005) 1173–1179. [DOI] [PubMed] [Google Scholar]
- [142].Liang J, Zhang Y, Xie T, Liu N, Chen H, Geng Y, Kurkciyan A, Mena JM, Stripp BR, Jiang D, Noble PW, Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice, Nature medicine, 22 (2016) 1285–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Boulay F, Tardif M, Brouchon L, Vignais P, Synthesis and use of a novel N-formyl peptide derivative to isolate a human N-formyl peptide receptor cDNA, Biochemical and biophysical research communications, 168 (1990) 1103–1109. [DOI] [PubMed] [Google Scholar]
- [144].Dorward DA, Lucas CD, Chapman GB, Haslett C, Dhaliwal K, Rossi AG, The role of formylated peptides and formyl peptide receptor 1 in governing neutrophil function during acute inflammation, The American journal of pathology, 185 (2015) 1172–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Martinez-Campos C, Burguete-Garcia AI, Madrid-Marina V, Role of TLR9 in Oncogenic Virus-Produced Cancer, Viral Immunol, 30 (2017) 98–105. [DOI] [PubMed] [Google Scholar]
- [146].Grazioli S, Pugin J, Mitochondrial Damage-Associated Molecular Patterns: From Inflammatory Signaling to Human Diseases, Front Immunol, 9 (2018) 832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ, Circulating mitochondrial DAMPs cause inflammatory responses to injury, Nature, 464 (2010) 104–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Scozzi D, Ibrahim M, Liao F, Lin X, Hsiao HM, Hachem R, Tague LK, Ricci A, Kulkarni HS, Huang HJ, Sugimoto S, Krupnick AS, Kreisel D, Gelman AE, Mitochondrial damage-associated molecular patterns released by lung transplants are associated with primary graft dysfunction, Am J Transplant, 19 (2019) 1464–1477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Han F, Wan S, Sun Q, Chen N, Li H, Zheng L, Zhang N, Huang Z, Hong L, Sun Q, Donor Plasma Mitochondrial DNA Is Correlated with Posttransplant Renal Allograft Function, Transplantation, 103 (2019) 2347–2358. [DOI] [PubMed] [Google Scholar]
- [150].Yousefi S, Mihalache C, Kozlowski E, Schmid I, Simon HU, Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps, Cell death and differentiation, 16 (2009) 1438–1444. [DOI] [PubMed] [Google Scholar]
- [151].Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, Malech HL, Ledbetter JA, Elkon KB, Kaplan MJ, Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease, Nat Med, 22 (2016) 146–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Sayah DM, Mallavia B, Liu F, Ortiz-Munoz G, Caudrillier A, DerHovanessian A, Ross DJ, Lynch JP 3rd, Saggar R, Ardehali A, Ware LB, Christie JD, Belperio JA, Looney MR, Neutrophil extracellular traps are pathogenic in primary graft dysfunction after lung transplantation, Am J Respir Crit Care Med, 191 (2015) 455–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Scozzi D, Wang X, Liao F, Liu Z, Zhu J, Pugh K, Ibrahim M, Hsiao HM, Miller MJ, Yizhan G, Mohanakumar T, Krupnick AS, Kreisel D, Gelman AE, Neutrophil extracellular trap fragments stimulate innate immune responses that prevent lung transplant tolerance, Am J Transplant, 19 (2019) 1011–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Mallavia B, Liu F, Lefrancais E, Cleary SJ, Kwaan N, Tian JJ, Magnen M, Sayah DM, Soong A, Chen J, Saggar R, Shino MY, Ross DJ, Derhovanessian A, Lynch Iii JP, Ardehali A, Weigt SS, Belperio JA, Hays SR, Golden JA, Leard LE, Shah RJ, Kleinhenz ME, Venado A, Kukreja J, Singer JP, Looney MR, Mitochondrial DNA Stimulates TLR9-Dependent NET Formation in Primary Graft Dysfunction, American journal of respiratory cell and molecular biology, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Honda M, Takeichi T, Hashimoto S, Yoshii D, Isono K, Hayashida S, Ohya Y, Yamamoto H, Sugawara Y, Inomata Y, Intravital Imaging of Neutrophil Recruitment Reveals the Efficacy of FPR1 Blockade in Hepatic Ischemia-Reperfusion Injury, J Immunol, 198 (2017) 1718–1728. [DOI] [PubMed] [Google Scholar]
- [156].Kharitonenkov A, Chen Z, Sures I, Wang H, Schilling J, Ullrich A, A family of proteins that inhibit signalling through tyrosine kinase receptors, Nature, 386 (1997) 181–186. [DOI] [PubMed] [Google Scholar]
- [157].Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, Lindberg FP, Role of CD47 as a marker of self on red blood cells, Science (New York, N.Y.), 288 (2000) 2051–2054. [DOI] [PubMed] [Google Scholar]
- [158].Okazawa H, Motegi S.-i., Ohyama N, Ohnishi H, Tomizawa T, Kaneko Y, Oldenborg P-A, Ishikawa O, Matozaki T, Negative Regulation of Phagocytosis in Macrophages by the CD47-SHPS-1 System, The Journal of Immunology, 174 (2005) 2004. [DOI] [PubMed] [Google Scholar]
- [159].Tsai RK, Discher DE, Inhibition of “self” engulfment through deactivation of myosin-II at the phagocytic synapse between human cells, The Journal of cell biology, 180 (2008) 989–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Lv Z, Bian Z, Shi L, Niu S, Ha B, Tremblay A, Li L, Zhang X, Paluszynski J, Liu M, Zen K, Liu Y, Loss of Cell Surface CD47 Clustering Formation and Binding Avidity to SIRPalpha Facilitate Apoptotic Cell Clearance by Macrophages, J Immunol, 195 (2015) 661–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Wang H, VerHalen J, Madariaga ML, Xiang S, Wang S, Lan P, Oldenborg PA, Sykes M, Yang YG, Attenuation of phagocytosis of xenogeneic cells by manipulating CD47, Blood, 109 (2007) 836–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Subramanian S, Parthasarathy R, Sen S, Boder ET, Discher DE, Species- and cell type-specific interactions between CD47 and human SIRPalpha, Blood, 107 (2006) 2548–2556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Subramanian S, Boder ET, Discher DE, Phylogenetic divergence of CD47 interactions with human signal regulatory protein alpha reveals locus of species specificity. Implications for the binding site, The Journal of biological chemistry, 282 (2007) 1805–1818. [DOI] [PubMed] [Google Scholar]
- [164].Takenaka K, Prasolava TK, Wang JC, Mortin-Toth SM, Khalouei S, Gan OI, Dick JE, Danska JS, Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells, Nat Immunol, 8 (2007) 1313–1323. [DOI] [PubMed] [Google Scholar]
- [165].Tena A, Kurtz J, Leonard DA, Dobrinsky JR, Terlouw SL, Mtango N, Verstegen J, Germana S, Mallard C, Arn JS, Sachs DH, Hawley RJ, Transgenic expression of human CD47 markedly increases engraftment in a murine model of pig-to-human hematopoietic cell transplantation, Am J Transplant, 14 (2014) 2713–2722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Tena AA, Sachs DH, Mallard C, Yang YG, Tasaki M, Farkash E, Rosales IA, Colvin RB, Leonard DA, Hawley RJ, Prolonged Survival of Pig Skin on Baboons After Administration of Pig Cells Expressing Human CD47, Transplantation, 101 (2017) 316–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Watanabe H, Sahara H, Nomura S, Tanabe T, Ekanayake-Alper DK, Boyd LK, Louras NJ, Asfour A, Danton MA, Ho SH, Arn SJ, Hawley RJ, Shimizu A, Nagayasu T, Ayares D, Lorber MI, Sykes M, Sachs DH, Yamada K, GalT-KO pig lungs are highly susceptible to acute vascular rejection in baboons, which may be mitigated by transgenic expression of hCD47 on porcine blood vessels, Xenotransplantation, 25 (2018) e12391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Watanabe H, Ariyoshi Y, Pomposelli T, Takeuchi K, Ekanayake-Alper DK, Boyd LK, Arn SJ, Sahara H, Shimizu A, Ayares D, Lorber MI, Sykes M, Sachs DH, Yamada K, Intra-bone bone marrow transplantation from hCD47 transgenic pigs to baboons prolongs chimerism to >60 days and promotes increased porcine lung transplant survival, Xenotransplantation, (2019) e12552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Dai H, Friday AJ, Abou-Daya KI, Williams AL, Mortin-Toth S, Nicotra ML, Rothstein DM, Shlomchik WD, Matozaki T, Isenberg JS, Oberbarnscheidt MH, Danska JS, Lakkis FG, Donor SIRPalpha polymorphism modulates the innate immune response to allogeneic grafts, Science immunology, 2 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Rosenblum MD, Olasz E, Woodliff JE, Johnson BD, Konkol MC, Gerber KA, Orentas RJ, Sandford G, Truitt RL, CD200 is a novel p53-target gene involved in apoptosis-associated immune tolerance, Blood, 103 (2004) 2691–2698. [DOI] [PubMed] [Google Scholar]
- [171].Wright GJ, Cherwinski H, Foster-Cuevas M, Brooke G, Puklavec MJ, Bigler M, Song Y, Jenmalm M, Gorman D, McClanahan T, Liu MR, Brown MH, Sedgwick JD, Phillips JH, Barclay AN, Characterization of the CD200 receptor family in mice and humans and their interactions with CD200, J Immunol, 171 (2003) 3034–3046. [DOI] [PubMed] [Google Scholar]
- [172].Wright GJ, Puklavec MJ, Willis AC, Hoek RM, Sedgwick JD, Brown MH, Barclay AN, Lymphoid/neuronal cell surface OX2 glycoprotein recognizes a novel receptor on macrophages implicated in the control of their function, Immunity, 13 (2000) 233–242. [DOI] [PubMed] [Google Scholar]
- [173].Hoek RM, Ruuls SR, Murphy CA, Wright GJ, Goddard R, Zurawski SM, Blom B, Homola ME, Streit WJ, Brown MH, Barclay AN, Sedgwick JD, Down-regulation of the macrophage lineage through interaction with OX2 (CD200), Science (New York, N.Y.), 290 (2000) 1768–1771. [DOI] [PubMed] [Google Scholar]
- [174].Gorczynski RM, Cattral MS, Chen Z, Hu J, Lei J, Min WP, Yu G, Ni J, An immunoadhesin incorporating the molecule OX-2 is a potent immunosuppressant that prolongs allo- and xenograft survival, J Immunol, 163 (1999) 1654–1660. [PubMed] [Google Scholar]
- [175].Gorczynski R, Chen Z, Khatri I, Yu K, Long-Term Tolerance and Skin Allograft Survival in CD200tg Mice After Autologous Marrow Transplantation, Transplantation, 98 (2014) 1271–1278. [DOI] [PubMed] [Google Scholar]
- [176].Gorczynski L, Chen Z, Hu J, Kai Y, Lei J, Ramakrishna V, Gorczynski RM, Evidence that an OX-2-positive cell can inhibit the stimulation of type 1 cytokine production by bone marrow-derived B7–1 (and B7–2)-positive dendritic cells, J Immunol, 162 (1999) 774–781. [PubMed] [Google Scholar]
- [177].Yan JJ, Koo TY, Lee HS, Lee WB, Kang B, Lee JG, Jang JY, Fang T, Ryu JH, Ahn C, Kim SJ, Yang J, Role of Human CD200 Overexpression in Pig-to-Human Xenogeneic Immune Response Compared With Human CD47 Overexpression, Transplantation, 102 (2018) 406–416. [DOI] [PubMed] [Google Scholar]
- [178].Savill J, Hogg N, Haslett C, Macrophage vitronectin receptor, CD36, and thrombospondin cooperate in recognition of neutrophils undergoing programmed cell death, Chest, 99 (1991) 6S–7S. [DOI] [PubMed] [Google Scholar]
- [179].Greenberg ME, Sun M, Zhang R, Febbraio M, Silverstein R, Hazen SL, Oxidized phosphatidylserine-CD36 interactions play an essential role in macrophage-dependent phagocytosis of apoptotic cells, The Journal of experimental medicine, 203 (2006) 2613–2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Albert ML, Pearce SF, Francisco LM, Sauter B, Roy P, Silverstein RL, Bhardwaj N, Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes, The Journal of experimental medicine, 188 (1998) 1359–1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Perry JSA, Russler-Germain EV, Zhou YW, Purtha W, Cooper ML, Choi J, Schroeder MA, Salazar V, Egawa T, Lee BC, Abumrad NA, Kim BS, Anderson MS, DiPersio JF, Hsieh CS, Transfer of Cell-Surface Antigens by Scavenger Receptor CD36 Promotes Thymic Regulatory T Cell Receptor Repertoire Development and Allo-tolerance, Immunity, 48 (2018) 923–936 e924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Nakano T, Ishimoto Y, Kishino J, Umeda M, Inoue K, Nagata K, Ohashi K, Mizuno K, Arita H, Cell adhesion to phosphatidylserine mediated by a product of growth arrest-specific gene 6, The Journal of biological chemistry, 272 (1997) 29411–29414. [DOI] [PubMed] [Google Scholar]
- [183].Anderson HA, Maylock CA, Williams JA, Paweletz CP, Shu H, Shacter E, Serum-derived protein S binds to phosphatidylserine and stimulates the phagocytosis of apoptotic cells, Nat Immunol, 4 (2003) 87–91. [DOI] [PubMed] [Google Scholar]
- [184].Scott RS, McMahon EJ, Pop SM, Reap EA, Caricchio R, Cohen PL, Earp HS, Matsushima GK, Phagocytosis and clearance of apoptotic cells is mediated by MER, Nature, 411 (2001) 207–211. [DOI] [PubMed] [Google Scholar]
- [185].Tibrewal N, Wu Y, D’Mello V, Akakura R, George TC, Varnum B, Birge RB, Autophosphorylation docking site Tyr-867 in Mer receptor tyrosine kinase allows for dissociation of multiple signaling pathways for phagocytosis of apoptotic cells and down-modulation of lipopolysaccharide-inducible NF-kappaB transcriptional activation, The Journal of biological chemistry, 283 (2008) 3618–3627. [DOI] [PubMed] [Google Scholar]
- [186].Rothlin CV, Ghosh S, Zuniga EI, Oldstone MB, Lemke G, TAM receptors are pleiotropic inhibitors of the innate immune response, Cell, 131 (2007) 1124–1136. [DOI] [PubMed] [Google Scholar]
- [187].Lu Q, Lemke G, Homeostatic regulation of the immune system by receptor tyrosine kinases of the Tyro 3 family, Science (New York, N.Y.), 293 (2001) 306–311. [DOI] [PubMed] [Google Scholar]
- [188].Camenisch TD, Koller BH, Earp HS, Matsushima GK, A novel receptor tyrosine kinase, Mer, inhibits TNF-alpha production and lipopolysaccharide-induced endotoxic shock, J Immunol, 162 (1999) 3498–3503. [PubMed] [Google Scholar]
- [189].Kheradmand T, Wang S, Bryant J, Tasch JJ, Lerret N, Pothoven KL, Houlihan JL, Miller SD, Zhang ZJ, Luo X, Ethylenecarbodiimide-fixed donor splenocyte infusions differentially target direct and indirect pathways of allorecognition for induction of transplant tolerance, J Immunol, 189 (2012) 804–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Bryant J, Lerret NM, Wang JJ, Kang HK, Tasch J, Zhang Z, Luo X, Preemptive donor apoptotic cell infusions induce IFN-gamma-producing myeloid-derived suppressor cells for cardiac allograft protection, J Immunol, 192 (2014) 6092–6101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Denney HA, Whittle RJ, Lai J, Jacques RM, Taylor PC, Regulatory T Cells in Chronic Graft-Versus-Host Disease After Extracorporeal Photopheresis: Correlation With Skin and Global Organ Responses, and Ability to Taper Steroids, Transplantation, 101 (2017) 204–211. [DOI] [PubMed] [Google Scholar]
- [192].Hachem R, Corris P, Extracorporeal Photopheresis for Bronchiolitis Obliterans Syndrome After Lung Transplantation, Transplantation, 102 (2018) 1059–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Yoo EK, Rook AH, Elenitsas R, Gasparro FP, Vowels BR, Apoptosis induction of ultraviolet light A and photochemotherapy in cutaneous T-cell Lymphoma: relevance to mechanism of therapeutic action, J Invest Dermatol, 107 (1996) 235–242. [DOI] [PubMed] [Google Scholar]
- [194].Mevorach D, Zuckerman T, Reiner I, Shimoni A, Samuel S, Nagler A, Rowe JM, Or R, Single infusion of donor mononuclear early apoptotic cells as prophylaxis for graft-versus-host disease in myeloablative HLA-matched allogeneic bone marrow transplantation: a phase I/IIa clinical trial, Biol Blood Marrow Transplant, 20 (2014) 58–65. [DOI] [PubMed] [Google Scholar]
- [195].Zhang L, DeBerge M, Wang J, Dangi A, Zhang X, Schroth S, Zhang Z, Thorp EB, Luo X, Receptor tyrosine kinase MerTK suppresses an allogenic type I IFN response to promote transplant tolerance, Am J Transplant, 19 (2019) 674–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Souza DG, Amaral FA, Fagundes CT, Coelho FM, Arantes RM, Sousa LP, Matzuk MM, Garlanda C, Mantovani A, Dias AA, Teixeira MM, The long pentraxin PTX3 is crucial for tissue inflammation after intestinal ischemia and reperfusion in mice, The American journal of pathology, 174 (2009) 1309–1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [197].Moller-Kristensen M, Wang W, Ruseva M, Thiel S, Nielsen S, Takahashi K, Shi L, Ezekowitz A, Jensenius JC, Gadjeva M, Mannan-binding lectin recognizes structures on ischaemic reperfused mouse kidneys and is implicated in tissue injury, Scandinavian journal of immunology, 61 (2005) 426–434. [DOI] [PubMed] [Google Scholar]
- [198].de Vries B, Walter SJ, Peutz-Kootstra CJ, Wolfs TG, van Heurn LW, Buurman WA, The mannose-binding lectin-pathway is involved in complement activation in the course of renal ischemiareperfusion injury, The American journal of pathology, 165 (2004) 1677–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [199].Hart ML, Ceonzo KA, Shaffer LA, Takahashi K, Rother RP, Reenstra WR, Buras JA, Stahl GL, Gastrointestinal ischemia-reperfusion injury is lectin complement pathway dependent without involving C1q, J Immunol, 174 (2005) 6373–6380. [DOI] [PubMed] [Google Scholar]
- [200].Bamboat ZM, Balachandran VP, Ocuin LM, Obaid H, Plitas G, DeMatteo RP, Toll-like receptor 9 inhibition confers protection from liver ischemia-reperfusion injury, Hepatology (Baltimore, Md.), 51 (2010) 621–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Han SJ, Li H, Kim M, Shlomchik MJ, Lee HT, Kidney Proximal Tubular TLR9 Exacerbates Ischemic Acute Kidney Injury, J Immunol, 201 (2018) 1073–1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Kitazume-Taneike R, Taneike M, Omiya S, Misaka T, Nishida K, Yamaguchi O, Akira S, Shattock MJ, Sakata Y, Otsu K, Ablation of Toll-like receptor 9 attenuates myocardial ischemia/reperfusion injury in mice, Biochemical and biophysical research communications, 515 (2019) 442–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Zhou QL, Teng F, Zhang YS, Sun Q, Cao YX, Meng GW, FPR1 gene silencing suppresses cardiomyocyte apoptosis and ventricular remodeling in rats with ischemia/reperfusion injury through the inhibition of MAPK signaling pathway, Experimental cell research, 370 (2018) 506–518. [DOI] [PubMed] [Google Scholar]