Abstract
While T cells play a critical role in protective immunity against infection, they are also responsible for graft rejection in the setting of transplantation. T cell differentiation is regulated by both intrinsic transcriptional pathways as well as extrinsic factors such as antigen encounter and the cytokine milieu. Herein, we review recent discoveries in the transcriptional regulation of T cell differentiation and their impact on the field of transplantation. Recent studies uncovering context-dependent differentiation programs that differ in the setting of infection or transplantation will also be discussed. Understanding the key transcriptional pathways that underlie T cell responses in transplantation has important clinical implications, including development of novel therapeutic agents to mitigate graft rejection.
Introduction
T cells are critical for protective immunity and for tumor surveillance, however, T cell responses against alloantigens are detrimental in the context of transplantation and are associated with graft rejection. T cell differentiation has been shown to be regulated by both cell-intrinsic transcriptional networks initiated following brief antigenic stimulation and also by cytokines in the environment. In this review we will focus on the transcriptional networks that regulate distinct programs of gene expression that control downstream effector or memory differentiation and function. Understanding these transcriptional pathways that program their development will facilitate the development of novel therapies to target these donor-reactive T cell populations.
Models of infection have shaped the majority of our understanding of programmed memory T cell differentiation, however recent reports have suggested that context-dependent programs exist, such that all of these lessons from models of microbial infection may not be applicable to the alloimmune response. This review will therefore focus on recent discoveries in the transcriptional regulation of T cell differentiation, specifically interferon regulatory factor 4 (IRF4), transcription factors associated with Th1 differentiation (Tbet and Eomesodermin) and the transcriptional regulation of tissue-resident memory cells (Trm). The impact of these results on the field of transplantation, as well as instances in which model-specific differences exist will also be discussed.
IRF4: A potent regulator of CD4+ T cell differentiation
Transplant-specific role for IRF4 in restricting T cell exhaustion
Since its essential role in lymphocyte biology was discovered 20 years ago [1], IRF4 (Interferon regulatory factor 4) has been primarily implicated in mediating the development and function of CD4+ T cell subsets, including Th2 [2–4], T follicular helper (Tfh) [5], Th9 [6–8], Th17 [9–11] and T regulatory cells (Tregs) [12, 13]. However, IRF4 is also a central regulator of T helper 1 (Th1) responses. Among CD4+ T cell subsets, alloreactive Th1 cells have been shown to cause allograft damage directly through Fas-Fas ligand–mediated cytotoxicity, or indirectly through the promotion of cytotoxic CD8+ T cells [14]. Like CD8+ T cells in chronic infection, graft-infiltrating CD4+ T cells express high amounts of IRF4 [15].
It has recently been demonstrated that IRF4 deficient CD4+ T cells showed impaired Th1 differentiation after in vitro and in vivo activation and that this was linked to impaired aerobic glycolysis and oxidative phosphorylation in the IRF4 deficient cells compared to controls [16, 17]. This finding was shown to be functionally relevant in a model of murine heterotopic cardiac transplantation by Wu et al. [15], where fully MHC-mismatched heart allografts survived indefinitely in mice where IRF4 was deleted conditionally in CD4+ T cells and where no other immunosuppressive therapies were administered. Interestingly, only IRF4, and not BATF- or STAT3-deficient animals, permanently accepted fully allogeneic Balb/c hearts [15] which is exceptional as there are very few single gene knockouts in which the rejection of fully allogeneic transplants is completely prevented. Of note, CD4+ and not CD8+ T cells are necessary and sufficient for allograft rejection in this model.
Of particular interest was the mechanism by which these animals became tolerant. It was not simply a matter of fewer allo-specific CD4+ T cells, nor was it Treg-mediated, as IRF4-deficient Treg cells are actually less suppressive than wild-type Tregs and their depletion does not result in rejection [15]. Tolerance was achieved in this setting through an IRF4-mediated repression of the expression of essential molecules associated with T cell dysfunction, namely PD-1 [15]. T cell dysfunction, such as exhaustion and anergy, represents a distinct T cell differentiation state following antigen encounter [18] and results in the induction of essential negative regulators that inhibit T cell function, like PD-1, amongst others [19]. This report of IRF4-mediated repression of T cell exhaustion is consistent with previous studies where IRF4-deficient T cells exhibited severe functional defects in T cell–mediated responses to microbial infection, allergy, graft-versus-host reaction, and autoimmunity [1, 6, 9, 20].
However, IRF4 has also been shown to drive the differentiation of cytotoxic effector CD8+ T cells [20–24]. Like in CD4+ T cells, IRF4 controls the expression of transcription factors involved in CD8+ effector cell differentiation including BLIMP1, Runx3, T-bet and TCF-1, as well as effector proteins such as cytokines and cytolytic proteins [23–25]. IRF4 is also involved in the metabolic changes of CD8+ T cells following activation. Dynamic reprogramming of metabolism is essential for T cell effector function and memory formation and compromised metabolic fitness has been closely linked to the functional impairments of exhausted T cells. IRF4 expression levels correlate with the strength of the TCR signal, thereby linking TCR affinity with the extent of metabolic changes following CD8+ T cell activation [25]. Interestingly, PD-1 signaling has also been shown to directly contribute to establishing the metabolic impairments of exhausted T cells [26], suggesting that exhaustion is underpinned by substantial rewiring of TCR signaling-mediated metabolic processes.
Interestingly, in a companion paper to Wu et al. [15], and in contrast to their findings for CD4+ T cells in transplantation, Man and colleagues [27] showed that while IRF4 was required for the initial expansion and subsequent maintenance of antigen-specific CD8+ T cells, high amounts of IRF4 promoted the establishment of T cell exhaustion in antigen-specific CD8+ T cells during chronic viral infection, including expression of the inhibitory receptors PD-1, TIM-3, and LAG-3, as well as altered cellular metabolism, and impaired cytokine secretion compared to fully functional effector and memory T cells [27].
Furthermore, they showed that IRF4, BATF, and NFAT (another TCR signaling-induced transcription factor) form a triplet that directly binds to and represses the expression of Tcf7, which encodes TCF1, and plays a critical role in the establishment of long-term memory T cells [28–32] thereby creating a self-reinforcing transcriptional circuit that establishes a network which promotes T cell exhaustion during chronic infection [27]. Importantly, reducing the dosage of IRF4 was sufficient to decrease expression of PD-1, increase anabolic metabolism, and partially restore secretion of IFN-γ, as well as release Tcf7 from repression, thus allowing for the development of memory-like T cells despite persistent viremia during chronic infection [27].
What might lead to such a dramatic model-specific and/or cell-specific difference in function for IRF4? There are several potential explanations. First, analysis of publically available genome-wide binding data shows that IRF4, BATF, and NFATc2 are often recruited to adjacent binding sites and preferentially co-occupy exhaustion-specific genes in CD8+ T cells, such as Pdcd-1 (encoding PD-1) [27]. In contrast, no evidence for this association was found in wild-type CD4+ T cells [15]. Rather, increased PD-1 expression correlated with the upregulation and binding of Helios (Ikzf2) to the Pdcd-1 locus and forced Helios expression promoted PD-1 expression in activated wild-type CD4+ T cells and since IRF4 represses Helios, PD-1 expression is not induced in activated wild-type CD4+ T cells [15]. Indeed, Helios expression is a feature of exhausted CD4+ but not CD8+ T cells [33].
Importantly, the initial dysfunctional state described on CD4+ Th1 cells by Wu et al. [15] in the setting of transplantation was shown to be reversible via blockade of the PD-1/PD-L1 signaling pathway during the initial days post-transplant, however it progressively evolved into a terminal irreversible state within 30 days post-transplant [15, 34]. Given this, as well as the recent reports describing graft rejection in stable transplant patients who were treated with PD-1 blockade for subsequent metastatic cancer [35, 36], pharmaceutical inhibition of IRF4 could be a potential novel therapeutic approach for transplantation.
Indeed, Wu et al. [15] found that the FDA approved MEK inhibitor, trametinib, effectively diminished IRF4 expression on activated CD4+ T cells and transient trametinib treatment prolonged allograft survival [15] further suggesting that targeting IRF4 may be a promising new therapy in the setting of transplantation. However, given the findings of Man et al. [27], the effects of IRF4 depletion therapy on CD8-mediated protective T cell responses should be closely monitored.
IRF4 is critical for IL-10 mediated Tr1 suppressive function
Recently, an additional role for IRF4 has been discovered in a subset of CD4+ regulatory T cells that are Foxp3 negative [4, 37, 38]. These cells, termed Type 1 regulatory (Tr1) cells, are driven by IL-27 and exert their immune regulation via secretion of IL-10 [39–43]. Although much is known about the development and function of traditional Treg cells, much less is known about the Tr1 subset, making the connection to the role of IRF4 in their development all the more compelling.
Interestingly, high spontaneous production of IL-10 or a polymorphism in the IL-10–592A allele has been observed to provide bone marrow transplant recipients with more favorable outcomes and fewer complications [44–47]. These observations led to a good deal of work to determine whether this alternative regulatory mechanism may play a role in promoting transplant tolerance. For instance, it has been shown that either through the adoptive transfer of IL-10 producing, Ag-specific Tr1 cells [48], or by pharmacological treatment with rapamycin plus IL-10, that active antigen-specific, long-term tolerance can be achieved in a murine model of allogeneic islet transplant via induction of Tr1 cells [49].
Tr1 cells may also play an important role in inducing transplantation tolerance in humans, as peripheral blood mononuclear cells (PBMC) isolated from patients who underwent islet transplant and became insulin independent produced significantly more IL-10 when compared with transplant subjects that continued to be insulin-dependent [47] and patients who spontaneously developed tolerance to kidney or liver grafts were shown to have an increased induction of Tr1 cells [50]. Taken together, these data indicate that IL-10-producing Tr1 cells may be naturally involved in inducing tolerance to allotransplants and targeting this population for expansion has potential to be therapeutic.
Specifically, Huang et al. [37] recently found that the Ras/MAPK pathways, downstream of T cell receptor (TCR) and IL-2 inducible T cell kinase (ITK) signaling, leads to IRF4 expression during Tr1 cell differentiation and that this expression is critical for the suppressive function of Tr1 cells [37]. These findings add to the reports of coordinated signaling of ITK and IRF4 in regulating Th9 cell differentiation [51]. Additionally, around the same time, a second group discovered that an IRF4-aryl hydrocarbon receptor (AhR)-dependent transcriptional network, induced by the cytokine Activin-A, also generates suppressive human Tr1 cells [38]. This was found to be due to IRF4 and AhR binding to IL-10 and ICOS promoter elements that controlled gene expression in human CD4+ T cells [38]. Indeed, silencing IRF4 abrogated the Activin-A–driven IL10 and ICOS up-regulation and thus impaired the suppressive functions of human Tr1 cells [38].
Since the ex vivo generation and expansion of Ag-specific nTreg cells remains a major challenge, the use of Tr1 cells, which are inducible Tregs and therefore able to be more easily generated in vitro in the presence of high levels of IL-10 and TCR stimulation [48], or by molecular manipulation [52] to be of the desired Ag specificity, the use of these cells for immunotherapy is a promising avenue for transplantation. By understanding the programmed development of Tr1 cells will facilitate the development of immunotherapies targeting this population to promote expansion and increased suppressor function in vivo.
T-bet and Eomesodermin: New mechanisms of old tricks
Active repression of non-Th1 CD4+ subsets by T-bet
It has recently come to light that T-bet plays an active role in repressing other CD4+ subsets via suppression of the Th17 master transcription factor RORγt [53]. This role for T-bet is of clinical relevance since in transplantation a role for Th17-mediated rejection has recently become appreciated. For instance, Yuan et al. [54] and Burrell et al. [55] have reported that Tbet deficient animals exhibited accelerated rejection associated with infiltration by predominantly IL-17–producing CD4 or CD8 T cells. More directly, blockade of this pathway through the use of IL-17 neutralizing antibodies or of IL-17 or RORγt deficient mice has shown prolonged renal and heart allograft survival as well as decreased severity of graft-versus-host disease (GVHD) [54, 56, 57]. Finally, Th17-producing memory cells from renal transplant recipients were shown to express high levels of the coinhibitory receptor CTLA-4 and thus are uniquely sensitive to CTLA-4-mediated inhibition [58]. This characteristic makes these cells resistant to CTLA-4 Ig-based costimulation blockade therapy, which blocks CTLA-4 as well as the intended CD28 target. Indeed, patients who experienced rejection while on belatacept exhibited increased frequencies of circulating Th17 cells relative to patients who were stable on belatacept or those that experienced rejection on tacrolimus. [58].
In addition to Th17 cell differentiation, Amarnath et al. [59] and Wang et al. [60] have recently described critical roles for T-bet in the regulation of Foxp3 expression in Tregs and for optimal T follicular helper (Tfh) cell responses, respectively. Th1, Th2, and Th17 cells have been shown to mediate acute GvHD syndromes, whereas Treg cells prevent acute GvHD and are associated with reduced clinical GvHD scores [59]. However, chronic GvHD is distinct from acute GvHD due to an autoimmune mechanism that is propagated by donor T cells that recognize host peptides presented by donor antigen-presenting cells [59]. Animal models have shown that a decrease in Treg cell numbers occurs along with an expansion of Th1 and Th17 cells leading to autoimmune-like pathology. Tbet has been shown to be vital for the development of acute GvHD [57], while its role during chronic GvHD is less clear.
Using a murine chronic GvHD model of alloreactive T-cell transfer, Amarnath and colleagues [59] found that the ability of transferred cells to secrete IFNγ did not affect the disease course, due to an equivalent ability to proliferate and traffic to target organs, even in the absence of IFNγ signaling [59]. Besancon et al. [61] have published similar findings in that T-bet, which is highly expressed in CD4+ and CD8+ graft-infiltrating T cells, but not IFNγ, is necessary for the development of an alloimmune response against fully mismatched pancreatic islet allografts [61]. Indeed, T-bet deficient mice permanently accepted allogeneic islets in 100% of cases and this correlated with a dramatic reduction of T cells expressing perforin, GzmB, and FasL and with their ability to produce IFNγ and TNFα [61]. However, in the absence of IFNγ signaling and deletion of T-bet, they found a cumulative increase in Treg cell numbers both in the secondary lymphoid organs and in GvHD target tissues in T-bet deficient recipients, associated with the generation of peripheral Treg cells from CD4+ T cells that had been primed in an alloreactive environment and this correlated with decreased pathogenesis [59].
Mechanistically, they found that T-bet directly binds to the Foxp3 promoter, thereby identifying a novel regulatory mechanism by which T-bet modulates peripheral Treg generation [59]. These results suggest that adoptive Treg cell therapy strategies currently being pursued to treat chronic GvHD may not be fully effective until methods to control Th1 signaling can be harnessed and the T-bet pathway therefore provides a feasible target in efforts to prevent chronic GvHD.
Importantly, taken together, this work suggests that by specifically inhibiting type II IFNγ receptor signaling, one might be able to augment Treg cells during chronic GvHD while maintaining type I mediated anti-viral responses. Thus, interventions that restrict the T-bet pathway might either be used alone or in combination with adoptive Treg-cell therapy for treatment of autoimmune chronic GvHD.
T-bet has also been shown to directly control the generation and function of Tfh in a context-dependent manner [60, 62]. As a CD4+ helper T cell subset specialized to aid in the germinal center (GC) reaction, Tfh cells have been reported to play important roles in many different types of immune responses [63–68]. Specifically, Wang et al. [60] found that while T-bet was dispensable for the early fate commitment of Tfh cells, it was essential for their maintenance by promoting their proliferation and inhibiting their apoptosis during acute viral infection [60]. Interestingly, T-bet deficient mice exhibited significant loss of Tfh cell responses during acute viral infection, but not in the setting of protein immunization [60], indicating a context dependent role for Tbet. These findings are of significant interest to the field of transplantation as a growing body of literature has uncovered an important role for Tfh cells in the generation of donor-specific antibody and subsequent allograft rejection [69–77]. Understanding the potential transplant-specific role for Tbet in promoting Tfh differentiation given these newly reported findings in the setting of infection and immunization may hold potential in developing Tfh-specific immunotherapies.
Complementary role for Eomesodermin in CD8+ T cell differentiation
Interestingly, it was noted early on that although CD4+ T cells and natural killer (NK) cells from T-bet deficient mice were defective in their ability to express IFNγ, CD8+ T cells could produce normal levels of IFNγ and had normal cytolytic function [78]. This implied a T-bet–independent pathway for effector differentiation of CD8+ T cells. Indeed, eomesodermin (Eomes), another T-box factor sharing 74% homology with T-bet and formally only known for its role in development, was found to be highly expressed in activated CD8+, but not CD4+ T cells in the absence of T-bet [79–81]. Further studies revealed that Eomes was co-expressed with perforin and GzmB on activated T cells indicating its likely role in invoking the cytolytic effector lineage. Thus, Eomes complements the role of T-bet in regulating cellular immunity by providing redundancy and acts cooperativity to induce T cell effector genes [79].
Since its discovery in T cells, much work has been done to further characterize the role of Eomes in CD8+ T cell differentiation [22, 79, 82–86]. Specifically, Banerjee et al. [82] observed a diminished capacity of CD8+ T cells lacking Eomes to compete within the Ag-specific memory population which led to defects in long-term persistence and secondary expansion post-rechallenge. These results suggest that Eomes confers competitive fitness to memory cells and supports a role for Eomes in promoting the persistence of long-lived memory (CD127hi KLRG-1lo) rather than terminal effector (CD127lo KLRG-1hi) cell differentiation of Ag-specific CD8+ T cells [82]. Further, Paley et al. [84] discovered that during chronic viral infections, Eomes expression in virus-specific CD8+ T cells was associated with markers of severe exhaustion and reduced co-production of antiviral cytokines.
While Th1 responses and T-bet expression have been linked to allograft rejection [87, 88] there is much less known about the role of Eomes in the regulation of alloimmunity. It has been shown that kidney transplant patients with primary cytomegalovirus (CMV) infection have increased expression of both T-bet and Eomes on CMV specific CD8+ T-cells [89]. Relatedly, we have recently identified a context-dependent differentiation program in which antigen-specific CD8+ T cells primed by a skin graft have increased recall potential and express less KLRG-1 and more CD127 compared to those primed by viral infection, which suggests that the T cell differentiation program induced in the context of transplantation is distinct from that induced in the context of exposure to a viral pathogen [90]. However, a specific role for Eomes in this dichotomy has not yet been established.
However, Ezzelarab and colleagues [85] recently observed that Eomes expression, but not T-bet expression, was upregulated on CD8+ T cells from transplant patients with stable graft function at 3-months post-transplant [85]. Interestingly, this high Eomes expression was correlated with an enhanced effector phenotype in allo-stimulated CD8+ T cells, independent of T-bet expression. Consistent with these findings, we have recently identified a subset of highly activated, effector CD8+ T cells that express the inhibitory Fc receptor FcγRIIB which preferentially inhibits CD44hiCD62Llo and Eomeslo cells [91].
Fcγ receptors play a pivotal role in immunity, controlling innate and humoral immunity by activating the effector functions of antibodies. Through both pharmacologic targeting of FcγRIIB, as well as CD8+ T cell-specific genetic deletion of Fcgr2b, we have shown that loss of this inhibitory molecule leads to the increased accumulation of CD8+ effector T cell resulting in accelerated graft rejection in mouse models. Moreover, FcγRIIB+ cells have lower transcripts of CD62L and Eomes, suggesting that FcγRIIB ligation may induce a negative signal in CD44hiCD62Llo and Eomeslo effector T cells that are induced to undergo apoptosis. Additionally, increased expression of FcγRIIB on CD8+ T cells correlated with freedom from rejection following withdrawal from immunosuppression in a clinical trial of kidney transplant recipients [91]. Because Eomes and CD62L are canonical molecules associated with high-quality memory T cell differentiation (reviewed in [92]), these data suggest that FcγRIIB functions to inhibit highly potent effector cells.
Regulation of T-bet by mTOR and implications for transplantation
Depending on the cytokine milieu available in the environment, CD4+ effector T cells differentiate into Th1, Th2, Th9, or Th17 subsets with different cytokine profiles and distinct functions. Th1 differentiation is initiated primarily through the induction of T-bet, a T-box transcription factor, leading to enhanced production of IFNγ [93]. For almost 20 years, much work has been conducted to understand the mechanisms by which T-bet regulates the expression of IFNγ, and thus drives differentiation of effector CD4+ and CD8+ T cells [83, 94–99].
mTOR serves as a core component of two distinct protein complexes, mTORC1 and mTORC2, which regulate different cellular processes [100, 101]. Recently, Chornoguz and colleagues [102] discovered 3 novel T-bet phosphorylation sites that were mTORC1 dependent, leading to T-bet’s ability to induce IFNγ production in CD4+ T cells [102]. One mechanism by which T-bet controls the expression of its target genes is by recruiting cellular machinery that delivers permissive and eliminates prohibitive epigenetic modifications. Consistent with this, simultaneous mutation of the three novel mTORC1-dependent phosphorylation sites discovered by Chornoguz et al. were found to reduce the T-bet mediated recruitment of chromatin remodeling complexes to the Ifng promoter [102]. These findings link mTOR-associated T cell activation with the induction of T-bet and production of IFNγ. Of note, these novel phosphorylation sites discovered in the mouse T-bet protein are also present in human T-bet [102].
Rapamycin, a macrolide antibiotic and immunosuppressive agent, has been clinically developed into two derivatives sirolimus and everolimus, which were approved by the Food and Drug Administration (FDA) for use in transplantation in 2001 and 2012, respectively [103]. Recently, intriguing data suggesting that a switch to, or the addition of, an mTOR inhibitor to the immunosuppressive regimen may aid in the successful management of posttransplant malignancies [104–106] which have historically been contraindicated for immune checkpoint inhibitor treatment, such as anti-PD-1 therapy, due to reports of upwards of 80% of patients experiencing rejection events and high overall patient mortality rates [36, 107, 108] when treated in this manner.
Specifically, Esfahani and colleagues [109], showed that pembrolizumab, an anti-PD-1 monoclonal antibody therapy, in combination with sirolimus led to the abatement of global T cell activation and proliferation, but maintained numbers of IFNγ-producing CD4+ T cells and cytotoxic CD8+ T cells within the circulation, thereby promoting a state of functional tolerance without a loss of immune-mediated anti-tumor activity [109]. Mechanistically, they showed that the eosinophilia-mediated allograft rejection associated with anti-PD-1 therapy [110–112] was brought under control by the addition of sirolimus [109], likely due to the effect of mTOR inhibition on T-bet-mediated recruitment of eosinophils to the graft [113–116], which can be linked to mTORc1 phosphorylation in light of Chornoguz et al.’s recent findings [102]. Interestingly, although not often discussed, eosinophilia-mediated rejection has long been noted clinically [117, 118], as has a role for Tbet expression in CD4+ T cells in the recruitment of eosinophils to the airways in the setting of asthma [113, 119], infection [114] and allergens [116]. It is interesting to speculate in light of the findings of Chornoguz et al. [102] that a similar mechanism is at play in the setting of transplantation.
It has long been thought that primarily Th1 effector CD4+ T cells as well as cytotoxic CD8+ T cells are required to mediate acute allograft rejection; however, a growing body of evidence suggests this is not the case. For instance, studies in a murine model of fully mismatched pancreatic islets have shown that IFNγ is not absolutely required for rejection of allogeneic islets or fully mismatched cardiac allografts; as IFNγ deficient mice were shown to rapidly reject their allografts [120–124]. Several studies have shown that this is directly linked to reduced T-bet expression, as T-bet deficiency contributed to the development of aggressive CD4+ and CD8+ Th17+ cell–mediated alloimmune responses that ultimately culminated in accelerated rejection that was costimulatory blockade resistant [54, 55, 58, 125, 126].
Moreover, mTOR has emerged as an important integrator of environmental cues which guide T cell effector and memory differentiation [86, 101, 127–130]. Interestingly, we have previously observed that blockade of the mTOR pathway, via exposure to rapamycin, paradoxically increases the quantity and quality of antigen-specific CD8+ T cell responses when the antigen is presented in the context of a viral infection, but fails to do so when the antigen is presented in the context of a transplant [131], consistent with other instances of context-dependent differentiation pathways.
Tissue-resident memory cells: Important passengers with implications for tolerance or rejection
Tissue-resident memory T (Trm) cells are a subset of long-lived memory T cells that are broadly distributed anatomically and occupy non-lymphoid tissues (NLT) without recirculating [132–137]. They are transcriptionally, phenotypically, and functionally distinct from recirculating central and effector memory T cells [138]. Their restricted anatomical localization in combination with their effector memory phenotype enables Trm cells to rapidly respond to local antigens [139]. However, these characteristics also make them potential mediators of both acute and chronic allograft rejection.
Trm are most commonly identified by their constitutive expression of CD69, which is induced upon migration to their site of residence where it antagonizes S1PR1-mediated tissue egress, allowing maintenance in NLT [138, 140–144]. However, recent evidence has shown that the functional requirement for CD69 is actually highly variable, depending on the tissue examined, playing no detectable role in generation of Trm at some sites and yet critical at others [145].
CD103, the receptor for the epithelial cell-specific ligand E-cadherin, is another universal marker of Trm cells and has long been known to endow peripheral CD8+ T cells with a unique capacity to access and damage the epithelial compartments of organ allografts [146–151]. More recently a direct link to a potential role for this molecule on Trm cells was made using transplant nephrectomies from immunosuppressed patients. Baan and colleagues [152] studied the presence, provenance (donor or recipient), and effector phenotype of CD103+ vs. CD103− CD69+ Trm cells and found that Trm cells are indeed present in the renal allograft, have strong immunostimulatory capacity, and finally, that the donor-derived cells present within the graft are mainly CD103+ Trm cells which are replaced by recipient-derived T cells within five months after transplantation [152].These data document a causal role for CD103+CD8+ T cells in promoting graft injury following allogeneic transplantation and suggest that targeting CD103+ T cells is a conceivable immunosuppressive target for combatting this highly cytolytic population.
The concept of donor-derived memory T cells present in, or being transferred along with the graft contributing to alloimmunity is not a new one [153–155] and yet still commands attention. A recent study showed that the presence of passenger CD4+ T cells in a donor cardiac graft enhanced cellular and humoral immune response in the recipient leading to acute graft rejection [156]. We and others have also shown that this augmentation is enhanced when donors are pre-sensitized to the recipient [156, 157]. Additionally, Zundler et al. [158] recently reported the accumulation of CD4+ Trm in the gut mucosa of patients with Inflammatory Bowel Disease (IBD) which predicted disease flares. Importantly, they found that depletion of this population led to the suppression of colitis activity, indicating the therapeuatic potential of targeting this population [158]. Indeed, ex vivo lung perfusion in the setting of transplantation has been shown to reduce donor leukocyte transfer into the recipient as well as decrease recipient T cell infiltration into lung allografts [159], indicating the potential to target depletion of Trm cells through perfusion. This is a promising therapeautic option, as the systemic administration of immunosuppressive agents has been shown to spare local tissue-resident T cell populations [160].
However, it is not a foregone conclusion that donor-derived memory T cells are bad and indeed several studies have shown a potentially beneficial role for this population [161–166]. Interestingly, it has recently been noted that the presence of Trm in lung and breast tumors [167–169], as well as in the livers of patients with chronic Hepatitis B Virus (HBV) infection [170], is associated with a better prognosis and better viral control, consistent with their important role in localized protective immunity. Sykes and colleagues [166, 171] have also shown T cell chimerism in the absence of graft-versus-host disease (GVHD) in the setting of human intestinal transplantation [166]. This chimerism led to two-way alloreactivity which dramatically impacted the rate of recipient T cell replacement in the graft mucosa, indicating the potential for donor-derived cells with graft specific TCR clones to have a positive effect by slowing down the threat of recipient-derived T cells and thus the development of rejection [171]. Thus, care and consideration must be taken in regards to depletion of this population given these data the potential exists that a rapid clearance of the donor-derived cells within the allograft may actually contribute to rejection [171].
In the setting of lung transplantation, periodic bronchoalveolar lavage (BAL) sampling of the airway enables prospective tracking of donor- and recipient-derived tissue resident T cells and the ability to potentially correlate tissue resident populations in patient BAL to clinical events [172]. Indeed, through a longitudinal single cell analysis of blood and BAL samples from more than 20 lung transplant recipients, Snyder et al. [172] was able to demonstrate through phenotypic, functional, and transcriptional analyses, that donor-derived T cells persist for more than one year post-transplantation in lung allografts as Trm T cells [172]. Rather surprisingly, they found that the long-term persistence of these donor lung Trm was associated with reduced incidence of clinical events that precipitate lung injury, including primary graft dysfunction, bacterial infection and acute cellular rejection [172].
This apparent discrepancy in the protective vs. pathogenic potential of Trm may be due to tissue specific Trm programing, which has recently been documented. For instance, compared to CD8+ Trm cells in the skin and intestine, lung CD8+ Trm cells are distinct at the transcriptional level [138], indicating specific adaptions to the local microenvironments.
Building off of their ground breaking 2013 paper in which they found a core Trm gene signature [138], Mackay et al. [173] further explored the genes encoding this signature and found the upregulation of Zfp683 (LOC100503878), or “homolog of Blimp1 in T cells” (Hobit), which is a transcription factor that had previously been identified in NKT cells [174]. Additionally, Hobit was specifically up-regulated in Trm cells and worked synergistically with Blimp1 to drive the establishment of a Trm population in the skin, gut, liver and kidney of mice by silencing lymphocyte egress from the tissues and by repressing the development of circulating memory cells [141, 173, 175]. Interestingly, Behr et al. [176] found that Blimp-1, but not Hobit, is essential for the formation of lung CD8+ Trm cells in the mouse. This finding highlights the unique transcriptional regulation of CD8+ Trm cells in different tissues, which may have implications for immunosuppressive strategies in solid organ transplantation.
Additionally, Hobit plays a particular role in maintaining the long-term cytolytic capabilities of CD8+ Trm cells within the tissue in both mice and humans [173, 177, 178] and Hobit rather than Blimp-1, is the essential transcriptional regulator in the long term maintenance of granzyme B-driven cytotoxicity of memory CD8+ T cells [179]. Specifically, the transcriptional regulation of Trm cytotoxicity appears to be temporally separated in CD8+ T cells with Blimp-1 initiating cytotoxic effector function and Hobit maintaining cytotoxicity in a deployment-ready state [179].
A third transcription factor, Runx3, has also been recently implicated in the differentiation and long-term maintenance of CD8+ Trm cells [180]. Runx3 is a well-established regulator of CD8+ T cell thymocyte development [181], and also supports cytotoxic activity of mature CD8+ T cells [182]. Using a combination of computative predictive algorithms, Milner et al. [180] was able to predict Trm regulatory transcription factors and confirm their function using an in vivo RNA interference (RNAi) loss-of-function screen. They evaluated both barrier and non-barrier Trm cells in order to reveal transcriptional regulators independent of tissue site. Runx3 was uncovered as a novel regulator of Trm differentiation and indeed, Runx3-deficiency resulted in a 50–150-fold loss of CD69+CD103+ Trm cells in both barrier and non-barrier tissues [180].
Temporally depleting Runx3 through tamoxifen administration further showed a crucial role for Runx3 not only in the differentiation, but also in the maintenance of Trm cell homeostasis [180]. Of particular interest, unlike Hobit and Blimp-1 [176], Runx3 was shown to be a universal regulator of Trm cell specification and was not tissue specific [180]. Lastly, similar to many of the other T cell specific differentiation programs, Runx3 was also shown to repress genes characteristic of circulating cells and promote those of tissue residency [180, 183].
Given these exciting new findings, and owing to the functional potential of Trm and their sheer abundance in various transplantable organs, they are likely a significant contributor to allogeneic immune responses and it remains to be seen whether Trm populations may hinder or help the promotion of transplant tolerance and may in fact depend on the specific tissue being transplanted.
In summary, the last few years have illuminated many novel T cell differentiation pathways and furthered our knowledge of previously existing pathways. These discoveries arm researchers with powerful new information to facilitate the development of novel therapies for the treatment and prevention of allograft rejection. However, several studies have also shown context-dependent differentiation pathways [60, 62, 90, 131], indicating that findings regarding T cell differentiation programs induced in the context of exposure to a viral pathogen may not hold true in the context of transplantation. Thus, the model in which scientific discoveries are made should be an important consideration in the development of novel therapies.
Concluding Remarks
It is now firmly established that alloreactive T cells accelerate graft rejection and prevent transplant tolerance. Despite decades of research on developing new immunosuppressive therapies with fewer toxicities, we have made little progress toward one transplant for life. The past several years has seen significant advances in our understanding of the transcriptional networks that drive T cell-intrinsic differentiation. Here we outline a few of these exciting new discoveries and discuss their relevance to the field of transplantation. We and others are also exploring many other intriguing lines of inquiry beyond the scope of this review, especially T follicular helper (Tfh) and T follicular regulatory (Tfr) cell differentiation and the interplay between FoxP3, STAT5, BCL6 and BLIMP 1 and the potential for targeting these pathways therapeautically to prevent the development of alloantibody or to prevent GVHD [69–71, 184–188]. Harnessing this information to develop novel therapeutics to promote regulatory T cell expansion or to divert harmful differentiation pathways could be a powerful new tool to prevent allograft rejection.
Highlights.
T cells play a critical role in protective immunity against infection, they are also responsible for graft rejection in the setting of transplantation.
T cell differentiation is regulated by both intrinsic transcriptional pathways as well as extrinsic factors such as antigen encounter and the cytokine milieu.
Herein, we review recent discoveries in the transcriptional regulation of T cell differentiation and their impact on the field of transplantation.
Recent studies uncovering context-dependent differentiation programs that differ in the setting of infection or transplantation will also be discussed.
Understanding the key transcriptional pathways that underlie T cell responses in transplantation has important clinical implications
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].Mittrucker HW, Matsuyama T, Grossman A, Kundig TM, Potter J, Shahinian A, Wakeham A, Patterson B, Ohashi PS, Mak TW, Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function, Science (New York, N.Y.), 275 (1997) 540–543. [DOI] [PubMed] [Google Scholar]
- [2].Lohoff M, Mittrucker HW, Prechtl S, Bischof S, Sommer F, Kock S, Ferrick DA, Duncan GS, Gessner A, Mak TW, Dysregulated T helper cell differentiation in the absence of interferon regulatory factor 4, Proc Natl Acad Sci U S A, 99 (2002) 11808–11812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Bruchard M, Rebe C, Derangere V, Togbe D, Ryffel B, Boidot R, Humblin E, Hamman A, Chalmin F, Berger H, Chevriaux A, Limagne E, Apetoh L, Vegran F, Ghiringhelli F, The receptor NLRP3 is a transcriptional regulator of TH2 differentiation, Nat Immunol, 16 (2015) 859–870. [DOI] [PubMed] [Google Scholar]
- [4].Ahyi AN, Chang HC, Dent AL, Nutt SL, Kaplan MH, IFN regulatory factor 4 regulates the expression of a subset of Th2 cytokines, J Immunol, 183 (2009) 1598–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Bollig N, Brustle A, Kellner K, Ackermann W, Abass E, Raifer H, Camara B, Brendel C, Giel G, Bothur E, Huber M, Paul C, Elli A, Kroczek RA, Nurieva R, Dong C, Jacob R, Mak TW, Lohoff M, Transcription factor IRF4 determines germinal center formation through follicular T-helper cell differentiation, Proc Natl Acad Sci U S A, 109 (2012) 8664–8669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Staudt V, Bothur E, Klein M, Lingnau K, Reuter S, Grebe N, Gerlitzki B, Hoffmann M, Ulges A, Taube C, Dehzad N, Becker M, Stassen M, Steinborn A, Lohoff M, Schild H, Schmitt E, Bopp T, Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells, Immunity, 33 (2010) 192–202. [DOI] [PubMed] [Google Scholar]
- [7].Jabeen R, Goswami R, Awe O, Kulkarni A, Nguyen ET, Attenasio A, Walsh D, Olson MR, Kim MH, Tepper RS, Sun J, Kim CH, Taparowsky EJ, Zhou B, Kaplan MH, Th9 cell development requires a BATF-regulated transcriptional network, J Clin Invest, 123 (2013) 4641–4653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Tamiya T, Ichiyama K, Kotani H, Fukaya T, Sekiya T, Shichita T, Honma K, Yui K, Matsuyama T, Nakao T, Fukuyama S, Inoue H, Nomura M, Yoshimura A, Smad2/3 and IRF4 play a cooperative role in IL-9-producing T cell induction, J Immunol, 191 (2013) 2360–2371. [DOI] [PubMed] [Google Scholar]
- [9].Brustle A, Heink S, Huber M, Rosenplanter C, Stadelmann C, Yu P, Arpaia E, Mak TW, Kamradt T, Lohoff M, The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4, Nat Immunol, 8 (2007) 958–966. [DOI] [PubMed] [Google Scholar]
- [10].Huber M, Brustle A, Reinhard K, Guralnik A, Walter G, Mahiny A, von Low E, Lohoff M, IRF4 is essential for IL-21-mediated induction, amplification, and stabilization of the Th17 phenotype, Proc Natl Acad Sci U S A, 105 (2008) 20846–20851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Li P, Spolski R, Liao W, Wang L, Murphy TL, Murphy KM, Leonard WJ, BATF-JUN is critical for IRF4-mediated transcription in T cells, Nature, 490 (2012) 543–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Zheng Y, Chaudhry A, Kas A, deRoos P, Kim JM, Chu TT, Corcoran L, Treuting P, Klein U, Rudensky AY, Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses, Nature, 458 (2009) 351–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Cretney E, Xin A, Shi W, Minnich M, Masson F, Miasari M, Belz GT, Smyth GK, Busslinger M, Nutt SL, Kallies A, The transcription factors Blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells, Nat Immunol, 12 (2011) 304–311. [DOI] [PubMed] [Google Scholar]
- [14].Liu Z, Fan H, Jiang S, CD4(+) T-cell subsets in transplantation, Immunol Rev, 252 (2013) 183–191. [DOI] [PubMed] [Google Scholar]
- [15].Wu J, Zhang H, Shi X, Xiao X, Fan Y, Minze LJ, Wang J, Ghobrial RM, Xia J, Sciammas R, Li XC, Chen W, Ablation of Transcription Factor IRF4 Promotes Transplant Acceptance by Driving Allogenic CD4(+) T Cell Dysfunction, Immunity, 47 (2017) 1114–1128.e1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Mahnke J, Schumacher V, Ahrens S, Kading N, Feldhoff LM, Huber M, Rupp J, Raczkowski F, Mittrucker HW, Interferon Regulatory Factor 4 controls TH1 cell effector function and metabolism, Scientific reports, 6 (2016) 35521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Kratchmarov R, Nish SA, Lin WHW, Adams WC, Chen YH, Yen B, Rothman NJ, Klein U, Reiner SL, IRF4 Couples Anabolic Metabolism to Th1 Cell Fate Determination, Immunohorizons, 1 (2017) 156–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Schietinger A, Philip M, Krisnawan VE, Chiu EY, Delrow JJ, Basom RS, Lauer P, Brockstedt DG, Knoblaugh SE, Hammerling GJ, Schell TD, Garbi N, Greenberg PD, Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis, Immunity, 45 (2016) 389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Wherry EJ, T cell exhaustion, Nat Immunol, 12 (2011) 492–499. [DOI] [PubMed] [Google Scholar]
- [20].Grusdat M, McIlwain DR, Xu HC, Pozdeev VI, Knievel J, Crome SQ, Robert-Tissot C, Dress RJ, Pandyra AA, Speiser DE, Lang E, Maney SK, Elford AR, Hamilton SR, Scheu S, Pfeffer K, Bode J, Mittrucker HW, Lohoff M, Huber M, Haussinger D, Ohashi PS, Mak TW, Lang KS, Lang PA, IRF4 and BATF are critical for CD8(+) T-cell function following infection with LCMV, Cell Death Differ, 21 (2014) 1050–1060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Nayar R, Enos M, Prince A, Shin H, Hemmers S, Jiang JK, Klein U, Thomas CJ, Berg LJ, TCR signaling via Tec kinase ITK and interferon regulatory factor 4 (IRF4) regulates CD8+ T-cell differentiation, Proc Natl Acad Sci U S A, 109 (2012) E2794–2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Nayar R, Schutten E, Jangalwe S, Durost PA, Kenney LL, Conley JM, Daniels K, Brehm MA, Welsh RM, Berg LJ, IRF4 Regulates the Ratio of T-Bet to Eomesodermin in CD8+ T Cells Responding to Persistent LCMV Infection, PloS one, 10 (2015) e0144826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Yao S, Buzo BF, Pham D, Jiang L, Taparowsky EJ, Kaplan MH, Sun J, Interferon regulatory factor 4 sustains CD8(+) T cell expansion and effector differentiation, Immunity, 39 (2013) 833–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Raczkowski F, Ritter J, Heesch K, Schumacher V, Guralnik A, Hocker L, Raifer H, Klein M, Bopp T, Harb H, Kesper DA, Pfefferle PI, Grusdat M, Lang PA, Mittrucker HW, Huber M, The transcription factor Interferon Regulatory Factor 4 is required for the generation of protective effector CD8+ T cells, Proc Natl Acad Sci U S A, 110 (2013) 15019–15024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Man K, Miasari M, Shi W, Xin A, Henstridge DC, Preston S, Pellegrini M, Belz GT, Smyth GK, Febbraio MA, Nutt SL, Kallies A, The transcription factor IRF4 is essential for TCR affinity-mediated metabolic programming and clonal expansion of T cells, Nat Immunol, 14 (2013) 1155–1165. [DOI] [PubMed] [Google Scholar]
- [26].Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, Stelekati E, McLane LM, Paley MA, Delgoffe GM, Wherry EJ, Bioenergetic Insufficiencies Due to Metabolic Alterations Regulated by the Inhibitory Receptor PD-1 Are an Early Driver of CD8(+) T Cell Exhaustion, Immunity, 45 (2016) 358–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Man K, Gabriel SS, Liao Y, Gloury R, Preston S, Henstridge DC, Pellegrini M, Zehn D, Berberich-Siebelt F, Febbraio MA, Shi W, Kallies A, Transcription Factor IRF4 Promotes CD8(+) T Cell Exhaustion and Limits the Development of Memory-like T Cells during Chronic Infection, Immunity, 47 (2017) 1129–1141.e1125. [DOI] [PubMed] [Google Scholar]
- [28].Zhou X, Yu S, Zhao DM, Harty JT, Badovinac VP, Xue HH, Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1, Immunity, 33 (2010) 229–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Jeannet G, Boudousquie C, Gardiol N, Kang J, Huelsken J, Held W, Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell memory, Proc Natl Acad Sci U S A, 107 (2010) 9777–9782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete S, Danilo M, Alfei F, Hofmann M, Wieland D, Pradervand S, Thimme R, Zehn D, Held W, T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections, Immunity, 45 (2016) 415–427. [DOI] [PubMed] [Google Scholar]
- [31].Leong YA, Chen Y, Ong HS, Wu D, Man K, Deleage C, Minnich M, Meckiff BJ, Wei Y, Hou Z, Zotos D, Fenix KA, Atnerkar A, Preston S, Chipman JG, Beilman GJ, Allison CC, Sun L, Wang P, Xu J, Toe JG, Lu HK, Tao Y, Palendira U, Dent AL, Landay AL, Pellegrini M, Comerford I, McColl SR, Schacker TW, Long HM, Estes JD, Busslinger M, Belz GT, Lewin SR, Kallies A, Yu D, CXCR5(+) follicular cytotoxic T cells control viral infection in B cell follicles, Nat Immunol, 17 (2016) 1187–1196. [DOI] [PubMed] [Google Scholar]
- [32].Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Nasti TH, Sharpe AH, Freeman GJ, Germain RN, Nakaya HI, Xue HH, Ahmed R, Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy, Nature, 537 (2016) 417–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BE, Wherry EJ, Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection, Immunity, 40 (2014) 289–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Zhang H, Wu J, Zou D, Xiao X, Yan H, Li XC, Chen W, Ablation of interferon regulatory factor 4 in T cells induces “memory” of transplant tolerance that is irreversible by immune checkpoint blockade, Am J Transplant, 19 (2019) 884–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Alhamad T, Venkatachalam K, Linette GP, Brennan DC, Checkpoint Inhibitors in Kidney Transplant Recipients and the Potential Risk of Rejection, Am J Transplant, 16 (2016) 1332–1333. [DOI] [PubMed] [Google Scholar]
- [36].Lipson EJ, Bagnasco SM, Moore J Jr., Jang S, Patel MJ, Zachary AA, Pardoll DM, Taube JM, Drake CG, Tumor Regression and Allograft Rejection after Administration of Anti-PD-1, The New England journal of medicine, 374 (2016) 896–898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Huang W, Solouki S, Koylass N, Zheng SG, August A, ITK signalling via the Ras/IRF4 pathway regulates the development and function of Tr1 cells, Nature communications, 8 (2017) 15871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Tousa S, Semitekolou M, Morianos I, Banos A, Trochoutsou AI, Brodie TM, Poulos N, Samitas K, Kapasa M, Konstantopoulos D, Paraskevopoulos G, Gaga M, Hawrylowicz CM, Sallusto F, Xanthou G, Activin-A co-opts IRF4 and AhR signaling to induce human regulatory T cells that restrain asthmatic responses, Proc Natl Acad Sci U S A, 114 (2017) E2891–e2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Groux H, O’Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG, A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis, Nature, 389 (1997) 737–742. [DOI] [PubMed] [Google Scholar]
- [40].Pot C, Jin H, Awasthi A, Liu SM, Lai CY, Madan R, Sharpe AH, Karp CL, Miaw SC, Ho IC, Kuchroo VK, Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells, J Immunol, 183 (2009) 797–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Vieira PL, Christensen JR, Minaee S, O’Neill EJ, Barrat FJ, Boonstra A, Barthlott T, Stockinger B, Wraith DC, O’Garra A, IL-10-secreting regulatory T cells do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+CD25+ regulatory T cells, J Immunol, 172 (2004) 5986–5993. [DOI] [PubMed] [Google Scholar]
- [42].Murugaiyan G, Mittal A, Lopez-Diego R, Maier LM, Anderson DE, Weiner HL, IL-27 is a key regulator of IL-10 and IL-17 production by human CD4+ T cells, J Immunol, 183 (2009) 2435–2443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Awasthi A, Carrier Y, Peron JP, Bettelli E, Kamanaka M, Flavell RA, Kuchroo VK, Oukka M, Weiner HL, A dominant function for interleukin 27 in generating interleukin 10-producing anti-inflammatory T cells, Nat Immunol, 8 (2007) 1380–1389. [DOI] [PubMed] [Google Scholar]
- [44].Baker KS, Roncarolo MG, Peters C, Bigler M, DeFor T, Blazar BR, High spontaneous IL-10 production in unrelated bone marrow transplant recipients is associated with fewer transplant-related complications and early deaths, Bone marrow transplantation, 23 (1999) 1123–1129. [DOI] [PubMed] [Google Scholar]
- [45].Bacchetta R, Bigler M, Touraine JL, Parkman R, Tovo PA, Abrams J, de Waal Malefyt R, de Vries JE, Roncarolo MG, High levels of interleukin 10 production in vivo are associated with tolerance in SCID patients transplanted with HLA mismatched hematopoietic stem cells, J Exp Med, 179 (1994) 493–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Lin MT, Storer B, Martin PJ, Tseng LH, Gooley T, Chen PJ, Hansen JA, Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation, The New England journal of medicine, 349 (2003) 2201–2210. [DOI] [PubMed] [Google Scholar]
- [47].Huurman VA, Velthuis JH, Hilbrands R, Tree TI, Gillard P, van der Meer-Prins PM, Duinkerken G, Pinkse GG, Keymeulen B, Roelen DL, Claas FH, Pipeleers DG, Roep BO, Allograft-specific cytokine profiles associate with clinical outcome after islet cell transplantation, Am J Transplant, 9 (2009) 382–388. [DOI] [PubMed] [Google Scholar]
- [48].Gagliani N, Jofra T, Stabilini A, Valle A, Atkinson M, Roncarolo MG, Battaglia M, Antigen-specific dependence of Tr1-cell therapy in preclinical models of islet transplant, Diabetes, 59 (2010) 433–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Battaglia M, Stabilini A, Draghici E, Gregori S, Mocchetti C, Bonifacio E, Roncarolo MG, Rapamycin and interleukin-10 treatment induces T regulatory type 1 cells that mediate antigen-specific transplantation tolerance, Diabetes, 55 (2006) 40–49. [PubMed] [Google Scholar]
- [50].VanBuskirk AM, Burlingham WJ, Jankowska-Gan E, Chin T, Kusaka S, Geissler F, Pelletier RP, Orosz CG, Human allograft acceptance is associated with immune regulation, J Clin Invest, 106 (2000) 145–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Gomez-Rodriguez J, Meylan F, Handon R, Hayes ET, Anderson SM, Kirby MR, Siegel RM, Schwartzberg PL, Itk is required for Th9 differentiation via TCR-mediated induction of IL-2 and IRF4, Nature communications, 7 (2016) 10857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Ahangarani RR, Janssens W, VanderElst L, Carlier V, VandenDriessche T, Chuah M, Weynand B, Vanoirbeek JA, Jacquemin M, Saint-Remy JM, In vivo induction of type 1-like regulatory T cells using genetically modified B cells confers long-term IL-10-dependent antigen-specific unresponsiveness, J Immunol, 183 (2009) 8232–8243. [DOI] [PubMed] [Google Scholar]
- [53].Lazarevic V, Chen X, Shim JH, Hwang ES, Jang E, Bolm AN, Oukka M, Kuchroo VK, Glimcher LH, T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORgammat, Nat Immunol, 12 (2011) 96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Yuan X, Paez-Cortez J, Schmitt-Knosalla I, D’Addio F, Mfarrej B, Donnarumma M, Habicht A, Clarkson MR, Iacomini J, Glimcher LH, Sayegh MH, Ansari MJ, A novel role of CD4 Th17 cells in mediating cardiac allograft rejection and vasculopathy, J Exp Med, 205 (2008) 3133–3144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Burrell BE, Csencsits K, Lu G, Grabauskiene S, Bishop DK, CD8+ Th17 mediate costimulation blockade-resistant allograft rejection in T-bet-deficient mice, J Immunol, 181 (2008) 3906–3914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Kwan T, Chadban SJ, Ma J, Bao S, Alexander SI, Wu H, IL-17 deficiency attenuates allograft injury and prolongs survival in a murine model of fully MHC-mismatched renal allograft transplantation, Am J Transplant, 15 (2015) 1555–1567. [DOI] [PubMed] [Google Scholar]
- [57].Yu Y, Wang D, Liu C, Kaosaard K, Semple K, Anasetti C, Yu XZ, Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORgammat in mice, Blood, 118 (2011) 5011–5020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Krummey SM, Cheeseman JA, Conger JA, Jang PS, Mehta AK, Kirk AD, Larsen CP, Ford ML, High CTLA-4 expression on Th17 cells results in increased sensitivity to CTLA-4 coinhibition and resistance to belatacept, Am J Transplant, 14 (2014) 607–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Amarnath S, Laurence A, Zhu N, Cunha R, Eckhaus MA, Taylor S, Foley JE, Ghosh M, Felizardo TC, Fowler DH, Tbet is a critical modulator of FoxP3 expression in autoimmune graft-versus-host disease, Haematologica, 102 (2017) 1446–1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Wang P, Wang Y, Xie L, Xiao M, Wu J, Xu L, Bai Q, Hao Y, Huang Q, Chen X, He R, Li B, Yang S, Chen Y, Wu Y, Ye L, The Transcription Factor T-Bet Is Required for Optimal Type I Follicular Helper T Cell Maintenance During Acute Viral Infection, Front Immunol, 10 (2019) 606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Besancon A, Demir Z, Goncalves T, Valette F, Panafieu E, Chatenoud L, You S, Differential Impact of T-bet and IFNgamma on Pancreatic Islet Allograft Rejection, Transplantation, 102 (2018) 1496–1504. [DOI] [PubMed] [Google Scholar]
- [62].Sheikh AA, Cooper L, Feng M, Souza-Fonseca-Guimaraes F, Lafouresse F, Duckworth BC, Huntington ND, Moon JJ, Pellegrini M, Nutt SL, Belz GT, Good-Jacobson KL, Groom JR, Context-Dependent Role for T-bet in T Follicular Helper Differentiation and Germinal Center Function following Viral Infection, Cell Rep, 28 (2019) 1758–1772 e1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Fahey LM, Wilson EB, Elsaesser H, Fistonich CD, McGavern DB, Brooks DG, Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells, J Exp Med, 208 (2011) 987–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Harker JA, Lewis GM, Mack L, Zuniga EI, Late interleukin-6 escalates T follicular helper cell responses and controls a chronic viral infection, Science (New York, N.Y.), 334 (2011) 825–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Hale JS, Youngblood B, Latner DR, Mohammed AU, Ye L, Akondy RS, Wu T, Iyer SS, Ahmed R, Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection, Immunity, 38 (2013) 805–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Glatman Zaretsky A, Taylor JJ, King IL, Marshall FA, Mohrs M, Pearce EJ, T follicular helper cells differentiate from Th2 cells in response to helminth antigens, J Exp Med, 206 (2009) 991–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Ding R, Gao W, He Z, Wang H, Conrad CD, Dinney CM, Yuan X, Wu F, Ma L, Wu Z, Liang C, Overrepresentation of Th1- and Th17-like Follicular Helper T Cells in Coronary Artery Disease, J Cardiovasc Transl Res, 8 (2015) 503–505. [DOI] [PubMed] [Google Scholar]
- [68].Smith C, Buhlmann JE, Wang X, Bartlett A, Lim B, Barrington RA, CD275-Independent IL-17-Producing T Follicular Helper-like Cells in Lymphopenic Autoimmune-Prone Mice, J Immunol, 196 (2016) 4935–4946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Danger R, Chesneau M, Delbos F, Le Bot S, Kerleau C, Chenouard A, Ville S, Degauque N, Conchon S, Cesbron A, Giral M, Brouard S, CXCR5(+)PD1(+)ICOS(+) Circulating T Follicular Helpers Are Associated With de novo Donor-Specific Antibodies After Renal Transplantation, Front Immunol, 10 (2019) 2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].La Muraglia GM 2nd, Wagener ME, Ford ML, Badell IR, Circulating T follicular helper cells are a biomarker of humoral alloreactivity and predict donor-specific antibody formation after transplantation, Am J Transplant, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Cano-Romero FL, Laguna Goya R, Utrero-Rico A, Gomez-Massa E, Arroyo-Sanchez D, Suarez-Fernandez P, Lora D, Andres A, Castro-Panete MJ, Paz-Artal E, Longitudinal profile of circulating T follicular helper lymphocytes parallels anti-HLA sensitization in renal transplant recipients, Am J Transplant, 19 (2019) 89–97. [DOI] [PubMed] [Google Scholar]
- [72].Chhabra M, Alsughayyir J, Qureshi MS, Mallik M, Ali JM, Gamper I, Moseley EL, Peacock S, Kosmoliaptsis V, Goddard MJ, Linterman MA, Motallebzadeh R, Pettigrew GJ, Germinal Center Alloantibody Responses Mediate Progression of Chronic Allograft Injury, Front Immunol, 9 (2018) 3038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Chenouard A, Chesneau M, Bui Nguyen L, Le Bot S, Cadoux M, Dugast E, Paul C, Malard-Castagnet S, Ville S, Guerif P, Soulillou JP, Degauque N, Danger R, Giral M, Brouard S, Renal Operational Tolerance Is Associated With a Defect of Blood Tfh Cells That Exhibit Impaired B Cell Help, Am J Transplant, 17 (2017) 1490–1501. [DOI] [PubMed] [Google Scholar]
- [74].Leibler C, Thiolat A, Henique C, Samson C, Pilon C, Tamagne M, Pirenne F, Vingert B, Cohen JL, Grimbert P, Control of Humoral Response in Renal Transplantation by Belatacept Depends on a Direct Effect on B Cells and Impaired T Follicular Helper-B Cell Crosstalk, Journal of the American Society of Nephrology : JASN, 29 (2018) 1049–1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Liarski VM, Kaverina N, Chang A, Brandt D, Yanez D, Talasnik L, Carlesso G, Herbst R, Utset TO, Labno C, Peng Y, Jiang Y, Giger ML, Clark MR, Cell distance mapping identifies functional T follicular helper cells in inflamed human renal tissue, Sci Transl Med, 6 (2014) 230ra246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Viglietti D, Loupy A, Vernerey D, Bentlejewski C, Gosset C, Aubert O, Duong van Huyen JP, Jouven X, Legendre C, Glotz D, Zeevi A, Lefaucheur C, Value of Donor-Specific Anti-HLA Antibody Monitoring and Characterization for Risk Stratification of Kidney Allograft Loss, Journal of the American Society of Nephrology : JASN, 28 (2017) 702–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Shi J, Luo F, Shi Q, Xu X, He X, Xia Y, Increased circulating follicular helper T cells with decreased programmed death-1 in chronic renal allograft rejection, BMC Nephrol, 16 (2015) 182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Szabo SJ, Sullivan BM, Stemmann C, Satoskar AR, Sleckman BP, Glimcher LH, Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells, Science (New York, N.Y.), 295 (2002) 338–342. [DOI] [PubMed] [Google Scholar]
- [79].Pearce EL, Mullen AC, Martins GA, Krawczyk CM, Hutchins AS, Zediak VP, Banica M, DiCioccio CB, Gross DA, Mao CA, Shen H, Cereb N, Yang SY, Lindsten T, Rossant J, Hunter CA, Reiner SL, Control of effector CD8+ T cell function by the transcription factor Eomesodermin, Science (New York, N.Y.), 302 (2003) 1041–1043. [DOI] [PubMed] [Google Scholar]
- [80].McLane LM, Banerjee PP, Cosma GL, Makedonas G, Wherry EJ, Orange JS, Betts MR, Differential localization of T-bet and Eomes in CD8 T cell memory populations, J Immunol, 190 (2013) 3207–3215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Knox JJ, Cosma GL, Betts MR, McLane LM, Characterization of T-bet and eomes in peripheral human immune cells, Front Immunol, 5 (2014) 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Banerjee A, Gordon SM, Intlekofer AM, Paley MA, Mooney EC, Lindsten T, Wherry EJ, Reiner SL, Cutting edge: The transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche, J Immunol, 185 (2010) 4988–4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Intlekofer AM, Takemoto N, Wherry EJ, Longworth SA, Northrup JT, Palanivel VR, Mullen AC, Gasink CR, Kaech SM, Miller JD, Gapin L, Ryan K, Russ AP, Lindsten T, Orange JS, Goldrath AW, Ahmed R, Reiner SL, Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin, Nat Immunol, 6 (2005) 1236–1244. [DOI] [PubMed] [Google Scholar]
- [84].Paley MA, Kroy DC, Odorizzi PM, Johnnidis JB, Dolfi DV, Barnett BE, Bikoff EK, Robertson EJ, Lauer GM, Reiner SL, Wherry EJ, Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection, Science (New York, N.Y.), 338 (2012) 1220–1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Perez-Gutierrez A, Metes DM, Lu L, Hariharan S, Thomson AW, Ezzelarab MB, Characterization of eomesodermin and T-bet expression by allostimulated CD8(+) T cells of healthy volunteers and kidney transplant patients in relation to graft outcome, Clin Exp Immunol, 194 (2018) 259–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Rao RR, Li Q, Odunsi K, Shrikant PA, The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin, Immunity, 32 (2010) 67–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Sun Q, Cheng D, Zhang M, He Q, Chen Z, Liu Z, Predominance of intraglomerular T-bet or GATA3 may determine mechanism of transplant rejection, Journal of the American Society of Nephrology : JASN, 22 (2011) 246–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Homs S, Mansour H, Desvaux D, Diet C, Hazan M, Buchler M, Lebranchu Y, Buob D, Badoual C, Matignon M, Audard V, Lang P, Grimbert P, Predominant Th1 and cytotoxic phenotype in biopsies from renal transplant recipients with transplant glomerulopathy, Am J Transplant, 9 (2009) 1230–1236. [DOI] [PubMed] [Google Scholar]
- [89].Hertoghs KM, Moerland PD, van Stijn A, Remmerswaal EB, Yong SL, van de Berg PJ, van Ham SM, Baas F, ten Berge IJ, van Lier RA, Molecular profiling of cytomegalovirus-induced human CD8+ T cell differentiation, J Clin Invest, 120 (2010) 4077–4090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Bozeman AM, Laurie SJ, Haridas D, Wagener ME, Ford ML, Transplantation preferentially induces a KLRG-1(lo) CD127(hi) differentiation program in antigen-specific CD8(+) T cells, Transplant immunology, 50 (2018) 34–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Morris AB, Farley CR, Pinelli DF, Adams LE, Cragg MS, Boss JM, Scharer CD, Fribourg M, Cravedi P, Heeger PS, Ford ML, Signaling through the inhibitory Fc receptor FcgRIIB induces CD8 T cell apoptosis to limit T cell immunity, Immunity, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Kaech SM, Cui W, Transcriptional control of effector and memory CD8+ T cell differentiation, Nat Rev Immunol, 12 (2012) 749–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH, A novel transcription factor, T-bet, directs Th1 lineage commitment, Cell, 100 (2000) 655–669. [DOI] [PubMed] [Google Scholar]
- [94].Gokmen MR, Dong R, Kanhere A, Powell N, Perucha E, Jackson I, Howard JK, Hernandez-Fuentes M, Jenner RG, Lord GM, Genome-wide regulatory analysis reveals that T-bet controls Th17 lineage differentiation through direct suppression of IRF4, J Immunol, 191 (2013) 5925–5932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Sullivan BM, Juedes A, Szabo SJ, von Herrath M, Glimcher LH, Antigen-driven effector CD8 T cell function regulated by T-bet, Proc Natl Acad Sci U S A, 100 (2003) 15818–15823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Yang Y, Xu J, Niu Y, Bromberg JS, Ding Y, T-bet and eomesodermin play critical roles in directing T cell differentiation to Th1 versus Th17, J Immunol, 181 (2008) 8700–8710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Joshi NS, Cui W, Chandele A, Lee HK, Urso DR, Hagman J, Gapin L, Kaech SM, Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor, Immunity, 27 (2007) 281–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Lighvani AA, Frucht DM, Jankovic D, Yamane H, Aliberti J, Hissong BD, Nguyen BV, Gadina M, Sher A, Paul WE, O’Shea JJ, T-bet is rapidly induced by interferon-gamma in lymphoid and myeloid cells, Proc Natl Acad Sci U S A, 98 (2001) 15137–15142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Oestreich KJ, Huang AC, Weinmann AS, The lineage-defining factors T-bet and Bcl-6 collaborate to regulate Th1 gene expression patterns, J Exp Med, 208 (2011) 1001–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Fantus D, Rogers NM, Grahammer F, Huber TB, Thomson AW, Roles of mTOR complexes in the kidney: implications for renal disease and transplantation, Nature reviews. Nephrology, 12 (2016) 587–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD, The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2, Nat Immunol, 12 (2011) 295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Chornoguz O, Hagan RS, Haile A, Arwood ML, Gamper CJ, Banerjee A, Powell JD, mTORC1 Promotes T-bet Phosphorylation To Regulate Th1 Differentiation, J Immunol, 198 (2017) 3939–3948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Weiler N, Bilge N, Troetschler S, Vermehren J, Schnitzbauer AA, Herrmann E, Sarrazin C, Zeuzem S, Welker MW, Conversion From Sirolimus to Everolimus in Long-Term Liver Graft Recipients, Journal of clinical pharmacology, 57 (2017) 837–845. [DOI] [PubMed] [Google Scholar]
- [104].Barnett R, Barta VS, Jhaveri KD, Preserved Renal-Allograft Function and the PD-1 Pathway Inhibitor Nivolumab, The New England journal of medicine, 376 (2017) 191–192. [DOI] [PubMed] [Google Scholar]
- [105].Knoll GA, Kokolo MB, Mallick R, Beck A, Buenaventura CD, Ducharme R, Barsoum R, Bernasconi C, Blydt-Hansen TD, Ekberg H, Felipe CR, Firth J, Gallon L, Gelens M, Glotz D, Gossmann J, Guba M, Morsy AA, Salgo R, Scheuermann EH, Tedesco-Silva H, Vitko S, Watson C, Fergusson DA, Effect of sirolimus on malignancy and survival after kidney transplantation: systematic review and meta-analysis of individual patient data, Bmj, 349 (2014) g6679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Klintmalm GB, Saab S, Hong JC, Nashan B, The role of mammalian target of rapamycin inhibitors in the management of post-transplant malignancy, Clinical transplantation, 28 (2014) 635–648. [DOI] [PubMed] [Google Scholar]
- [107].Abdel-Wahab N, Safa H, Abudayyeh A, Johnson DH, Trinh VA, Zobniw CM, Lin H, Wong MK, Abdelrahim M, Gaber AO, Suarez-Almazor ME, Diab A, Checkpoint inhibitor therapy for cancer in solid organ transplantation recipients: an institutional experience and a systematic review of the literature, J Immunother Cancer, 7 (2019) 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Aguirre LE, Guzman ME, Lopes G, Hurley J, Immune Checkpoint Inhibitors and the Risk of Allograft Rejection: A Comprehensive Analysis on an Emerging Issue, Oncologist, 24 (2019) 394–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Esfahani K, Al-Aubodah TA, Thebault P, Lapointe R, Hudson M, Johnson NA, Baran D, Bhulaiga N, Takano T, Cailhier JF, Piccirillo CA, Miller WH, Targeting the mTOR pathway uncouples the efficacy and toxicity of PD-1 blockade in renal transplantation, Nature communications, 10 (2019) 4712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Bernard-Tessier A, Jeanville P, Champiat S, Lazarovici J, Voisin AL, Mateus C, Lambotte O, Annereau M, Michot JM, Immune-related eosinophilia induced by anti-programmed death 1 or death-ligand 1 antibodies, Eur J Cancer, 81 (2017) 135–137. [DOI] [PubMed] [Google Scholar]
- [111].Weir MR, Hall-Craggs M, Shen SY, Posner JN, Alongi SV, Dagher FJ, Sadler JH, The prognostic value of the eosinophil in acute renal allograft rejection, Transplantation, 41 (1986) 709–712. [DOI] [PubMed] [Google Scholar]
- [112].Braun MY, Desalle F, Le Moine A, Pretolani M, Matthys P, Kiss R, Goldman M, IL-5 and eosinophils mediate the rejection of fully histoincompatible vascularized cardiac allografts: regulatory role of alloreactive CD8(+) T lymphocytes and IFN-gamma, European journal of immunology, 30 (2000) 1290–1296. [DOI] [PubMed] [Google Scholar]
- [113].Fujiwara M, Hirose K, Kagami S, Takatori H, Wakashin H, Tamachi T, Watanabe N, Saito Y, Iwamoto I, Nakajima H, T-bet inhibits both TH2 cell-mediated eosinophil recruitment and TH17 cell-mediated neutrophil recruitment into the airways, The Journal of allergy and clinical immunology, 119 (2007) 662–670. [DOI] [PubMed] [Google Scholar]
- [114].Glanville N, Peel TJ, Schroder A, Aniscenko J, Walton RP, Finotto S, Johnston SL, Tbet Deficiency Causes T Helper Cell Dependent Airways Eosinophilia and Mucus Hypersecretion in Response to Rhinovirus Infection, PLoS pathogens, 12 (2016) e1005913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Hua W, Liu H, Xia LX, Tian BP, Huang HQ, Chen ZY, Ju ZY, Li W, Chen ZH, Shen HH, Rapamycin inhibition of eosinophil differentiation attenuates allergic airway inflammation in mice, Respirology, 20 (2015) 1055–1065. [DOI] [PubMed] [Google Scholar]
- [116].Matsuki A, Takatori H, Makita S, Yokota M, Tamachi T, Suto A, Suzuki K, Hirose K, Nakajima H, T-bet inhibits innate lymphoid cell-mediated eosinophilic airway inflammation by suppressing IL-9 production, The Journal of allergy and clinical immunology, 139 (2017) 1355–1367.e1356. [DOI] [PubMed] [Google Scholar]
- [117].Martinez OM, Ascher NL, Ferrell L, Villanueva J, Lake J, Roberts JP, Krams SM, Evidence for a nonclassical pathway of graft rejection involving interleukin 5 and eosinophils, Transplantation, 55 (1993) 909–918. [DOI] [PubMed] [Google Scholar]
- [118].Chan SY, DeBruyne LA, Goodman RE, Eichwald EJ, Bishop DK, In vivo depletion of CD8+ T cells results in Th2 cytokine production and alternate mechanisms of allograft rejection, Transplantation, 59 (1995) 1155–1161. [PubMed] [Google Scholar]
- [119].Finotto S, Neurath MF, Glickman JN, Qin S, Lehr HA, Green FH, Ackerman K, Haley K, Galle PR, Szabo SJ, Drazen JM, De Sanctis GT, Glimcher LH, Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet, Science (New York, N.Y.), 295 (2002) 336–338. [DOI] [PubMed] [Google Scholar]
- [120].Nicolls MR, Coulombe M, Diamond AS, Beilke J, Gill RG, Interferon-gamma is not a universal requirement for islet allograft survival, Transplantation, 74 (2002) 472–477. [DOI] [PubMed] [Google Scholar]
- [121].Saleem S, Konieczny BT, Lowry RP, Baddoura FK, Lakkis FG, Acute rejection of vascularized heart allografts in the absence of IFNgamma, Transplantation, 62 (1996) 1908–1911. [DOI] [PubMed] [Google Scholar]
- [122].Konieczny BT, Dai Z, Elwood ET, Saleem S, Linsley PS, Baddoura FK, Larsen CP, Pearson TC, Lakkis FG, IFN-gamma is critical for long-term allograft survival induced by blocking the CD28 and CD40 ligand T cell costimulation pathways, J Immunol, 160 (1998) 2059–2064. [PubMed] [Google Scholar]
- [123].Valujskikh A, Heeger PS, CD4+ T cells responsive through the indirect pathway can mediate skin graft rejection in the absence of interferon-gamma, Transplantation, 69 (2000) 1016–1019. [DOI] [PubMed] [Google Scholar]
- [124].Zhou P, Szot GL, Guo Z, Kim O, He G, Wang J, Grusby MJ, Newell KA, Thistlethwaite JR, Bluestone JA, Alegre ML, Role of STAT4 and STAT6 signaling in allograft rejection and CTLA4-Ig-mediated tolerance, J Immunol, 165 (2000) 5580–5587. [DOI] [PubMed] [Google Scholar]
- [125].Bishop DK, Chan Wood S, Eichwald EJ, Orosz CG, Immunobiology of allograft rejection in the absence of IFN-gamma: CD8+ effector cells develop independently of CD4+ cells and CD40-CD40 ligand interactions, J Immunol, 166 (2001) 3248–3255. [DOI] [PubMed] [Google Scholar]
- [126].Mathur AN, Chang HC, Zisoulis DG, Kapur R, Belladonna ML, Kansas GS, Kaplan MH, T-bet is a critical determinant in the instability of the IL-17-secreting T-helper phenotype, Blood, 108 (2006) 1595–1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R, mTOR regulates memory CD8 T-cell differentiation, Nature, 460 (2009) 108–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, Worley PF, Kozma SC, Powell JD, The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment, Immunity, 30 (2009) 832–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Pollizzi KN, Waickman AT, Patel CH, Sun IH, Powell JD, Cellular size as a means of tracking mTOR activity and cell fate of CD4+ T cells upon antigen recognition, PloS one, 10 (2015) e0121710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Zeiser R, Leveson-Gower DB, Zambricki EA, Kambham N, Beilhack A, Loh J, Hou JZ, Negrin RS, Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells, Blood, 111 (2008) 453–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Ferrer IR, Wagener ME, Robertson JM, Turner AP, Araki K, Ahmed R, Kirk AD, Larsen CP, Ford ML, Cutting edge: Rapamycin augments pathogen-specific but not graft-reactive CD8+ T cell responses, J Immunol, 185 (2010) 2004–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Steinert EM, Schenkel JM, Fraser KA, Beura LK, Manlove LS, Igyarto BZ, Southern PJ, Masopust D, Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance, Cell, 161 (2015) 737–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJ, Bickham KL, Lerner H, Goldstein M, Sykes M, Kato T, Farber DL, Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets, Immunity, 38 (2013) 187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Thome JJ, Yudanin N, Ohmura Y, Kubota M, Grinshpun B, Sathaliyawala T, Kato T, Lerner H, Shen Y, Farber DL, Spatial map of human T cell compartmentalization and maintenance over decades of life, Cell, 159 (2014) 814–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Masopust D, Vezys V, Marzo AL, Lefrancois L, Preferential localization of effector memory cells in nonlymphoid tissue, Science (New York, N.Y.), 291 (2001) 2413–2417. [DOI] [PubMed] [Google Scholar]
- [136].Hogan RJ, Zhong W, Usherwood EJ, Cookenham T, Roberts AD, Woodland DL, Protection from respiratory virus infections can be mediated by antigen-specific CD4(+) T cells that persist in the lungs, J Exp Med, 193 (2001) 981–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Reinhardt RL, Khoruts A, Merica R, Zell T, Jenkins MK, Visualizing the generation of memory CD4 T cells in the whole body, Nature, 410 (2001) 101–105. [DOI] [PubMed] [Google Scholar]
- [138].Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon M-L, Vega-Ramos J, Lauzurica P, Mueller SN, Stefanovic T, Tscharke DC, Heath WR, Inouye M, Carbone FR, Gebhardt T, The developmental pathway for CD103+CD8+ tissue-resident memory T cells of skin, Nature Immunology, 14 (2013) 1294–1301. [DOI] [PubMed] [Google Scholar]
- [139].Schenkel JM, Fraser KA, Vezys V, Masopust D, Sensing and alarm function of resident memory CD8(+) T cells, Nat Immunol, 14 (2013) 509–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Masopust D, Vezys V, Wherry EJ, Barber DL, Ahmed R, Cutting edge: gut microenvironment promotes differentiation of a unique memory CD8 T cell population, J Immunol, 176 (2006) 2079–2083. [DOI] [PubMed] [Google Scholar]
- [141].Shiow LR, Rosen DB, Brdickova N, Xu Y, An J, Lanier LL, Cyster JG, Matloubian M, CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs, Nature, 440 (2006) 540–544. [DOI] [PubMed] [Google Scholar]
- [142].Lee YT, Suarez-Ramirez JE, Wu T, Redman JM, Bouchard K, Hadley GA, Cauley LS, Environmental and antigen receptor-derived signals support sustained surveillance of the lungs by pathogen-specific cytotoxic T lymphocytes, Journal of virology, 85 (2011) 4085–4094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Mackay LK, Wynne-Jones E, Freestone D, Pellicci DG, Mielke LA, Newman DM, Braun A, Masson F, Kallies A, Belz GT, Carbone FR, T-box Transcription Factors Combine with the Cytokines TGF-beta and IL-15 to Control Tissue-Resident Memory T Cell Fate, Immunity, 43 (2015) 1101–1111. [DOI] [PubMed] [Google Scholar]
- [144].Kumar BV, Ma W, Miron M, Granot T, Guyer RS, Carpenter DJ, Senda T, Sun X, Ho SH, Lerner H, Friedman AL, Shen Y, Farber DL, Human Tissue-Resident Memory T Cells Are Defined by Core Transcriptional and Functional Signatures in Lymphoid and Mucosal Sites, Cell Rep, 20 (2017) 2921–2934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Walsh DA, Borges da Silva H, Beura LK, Peng C, Hamilton SE, Masopust D, Jameson SC, The Functional Requirement for CD69 in Establishment of Resident Memory CD8(+) T Cells Varies with Tissue Location, J Immunol, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Hadley GA, Bartlett ST, Via CS, Rostapshova EA, Moainie S, The epithelial cell-specific integrin, CD103 (alpha E integrin), defines a novel subset of alloreactive CD8+ CTL, J Immunol, 159 (1997) 3748–3756. [PubMed] [Google Scholar]
- [147].Feng Y, Wang D, Yuan R, Parker CM, Farber DL, Hadley GA, CD103 expression is required for destruction of pancreatic islet allografts by CD8(+) T cells, J Exp Med, 196 (2002) 877–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Ding R, Li B, Muthukumar T, Dadhania D, Medeiros M, Hartono C, Serur D, Seshan SV, Sharma VK, Kapur S, Suthanthiran M, CD103 mRNA levels in urinary cells predict acute rejection of renal allografts, Transplantation, 75 (2003) 1307–1312. [DOI] [PubMed] [Google Scholar]
- [149].Wang D, Yuan R, Feng Y, El-Asady R, Farber DL, Gress RE, Lucas PJ, Hadley GA, Regulation of CD103 expression by CD8+ T cells responding to renal allografts, J Immunol, 172 (2004) 214–221. [DOI] [PubMed] [Google Scholar]
- [150].Yuan R, El-Asady R, Liu K, Wang D, Drachenberg CB, Hadley GA, Critical role for CD103+CD8+ effectors in promoting tubular injury following allogeneic renal transplantation, J Immunol, 175 (2005) 2868–2879. [DOI] [PubMed] [Google Scholar]
- [151].Heeger PS, Greenspan NS, Kuhlenschmidt S, Dejelo C, Hricik DE, Schulak JA, Tary-Lehmann M, Pretransplant frequency of donor-specific, IFN-gamma-producing lymphocytes is a manifestation of immunologic memory and correlates with the risk of posttransplant rejection episodes, J Immunol, 163 (1999) 2267–2275. [PubMed] [Google Scholar]
- [152].de Leur K, Dieterich M, Hesselink DA, Corneth OBJ, Dor F, de Graav GN, Peeters AMA, Mulder A, Kimenai H, Claas FHJ, Clahsen-van Groningen MC, van der Laan LJW, Hendriks RW, Baan CC, Characterization of donor and recipient CD8+ tissue-resident memory T cells in transplant nephrectomies, Scientific reports, 9 (2019) 5984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Starzl TE, Demetris AJ, Trucco M, Murase N, Ricordi C, Ildstad S, Ramos H, Todo S, Tzakis A, Fung JJ, Nalesnik M, Zeevi A, Rudert WA, Kocova M, Cell Migration and Chimerism After Whole-organ Transplantation: The Basis of Graft Acceptance, Hepatology, 17 (1993) 1127–1152. [PMC free article] [PubMed] [Google Scholar]
- [154].Josien R, Heslan M, Brouard S, Soulillou JP, Cuturi MC, Critical requirement for graft passenger leukocytes in allograft tolerance induced by donor blood transfusion, Blood, 92 (1998) 4539–4544. [PubMed] [Google Scholar]
- [155].Schlitt HJ, Hundrieser J, Ringe B, Pichlmayr R, Donor-type microchimerism associated with graft rejection eight years after liver transplantation, The New England journal of medicine, 330 (1994) 646–647. [DOI] [PubMed] [Google Scholar]
- [156].Harper IG, Ali JM, Harper SJ, Wlodek E, Alsughayyir J, Negus MC, Qureshi MS, Motalleb-Zadeh R, Saeb-Parsy K, Bolton EM, Bradley JA, Clatworthy MR, Conlon TM, Pettigrew GJ, Augmentation of Recipient Adaptive Alloimmunity by Donor Passenger Lymphocytes within the Transplant, Cell Rep, 15 (2016) 1214–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Crepeau R, Role of Donor-Derived Tissue Resident Memory T cells in Modulating Alloimmunity. - ATC Abstracts, American Transplant CongressChicago, IL, 2017. [Google Scholar]
- [158].Zundler S, Becker E, Spocinska M, Slawik M, Parga-Vidal L, Stark R, Wiendl M, Atreya R, Rath T, Leppkes M, Hildner K, Lopez-Posadas R, Lukassen S, Ekici AB, Neufert C, Atreya I, van Gisbergen K, Neurath MF, Hobit- and Blimp-1-driven CD4(+) tissue-resident memory T cells control chronic intestinal inflammation, Nat Immunol, 20 (2019) 288–300. [DOI] [PubMed] [Google Scholar]
- [159].Stone JP, Critchley WR, Major T, Rajan G, Risnes I, Scott H, Liao Q, Wohlfart B, Sjoberg T, Yonan N, Steen S, Fildes JE, Altered Immunogenicity of Donor Lungs via Removal of Passenger Leukocytes Using Ex Vivo Lung Perfusion, Am J Transplant, 16 (2016) 33–43. [DOI] [PubMed] [Google Scholar]
- [160].Clark RA, Watanabe R, Teague JE, Schlapbach C, Tawa MC, Adams N, Dorosario AA, Chaney KS, Cutler CS, LeBoeuf NR, Carter JB, Fisher DC, Kupper TS, Skin Effector Memory T Cells Do Not Recirculate and Provide Immune Protection in Alemtuzumab-Treated CTCL Patients, (2012). [DOI] [PMC free article] [PubMed]
- [161].Starzl TE, Demetris AJ, Murase N, Ildstad S, Ricordi C, Trucco M, Cell migration, chimerism, and graft acceptance, Lancet (London, England), 339 (1992) 1579–1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Kawai T, Cosimi AB, Spitzer TR, Tolkoff-Rubin N, Suthanthiran M, Saidman SL, Shaffer J, Preffer FI, Ding R, Sharma V, Fishman JA, Dey B, Ko DS, Hertl M, Goes NB, Wong W, Williams WW Jr., Colvin RB, Sykes M, Sachs DH, HLA-mismatched renal transplantation without maintenance immunosuppression, The New England journal of medicine, 358 (2008) 353–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Nadazdin O, Boskovic S, Murakami T, Tocco G, Smith RN, Colvin RB, Sachs DH, Allan J, Madsen JC, Kawai T, Cosimi AB, Benichou G, Host alloreactive memory T cells influence tolerance to kidney allografts in nonhuman primates, Sci Transl Med, 3 (2011) 86ra51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Yamada Y, Boskovic S, Aoyama A, Murakami T, Putheti P, Smith RN, Ochiai T, Nadazdin O, Koyama I, Boenisch O, Najafian N, Bhasin MK, Colvin RB, Madsen JC, Strom TB, Sachs DH, Benichou G, Cosimi AB, Kawai T, Overcoming memory T-cell responses for induction of delayed tolerance in nonhuman primates, Am J Transplant, 12 (2012) 330–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Yamada Y, Ochiai T, Boskovic S, Nadazdin O, Oura T, Schoenfeld D, Cappetta K, Smith RN, Colvin RB, Madsen JC, Sachs DH, Benichou G, Cosimi AB, Kawai T, Use of CTLA4Ig for induction of mixed chimerism and renal allograft tolerance in nonhuman primates, Am J Transplant, 14 (2014) 2704–2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Zuber J, Rosen S, Shonts B, Sprangers B, Savage TM, Richman S, Yang S, Lau SP, DeWolf S, Farber D, Vlad G, Zorn E, Wong W, Emond J, Levin B, Martinez M, Kato T, Sykes M, Macrochimerism in Intestinal Transplantation: Association With Lower Rejection Rates and Multivisceral Transplants, Without GVHD, Am J Transplant, 15 (2015) 2691–2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Ganesan AP, Clarke J, Wood O, Garrido-Martin EM, Chee SJ, Mellows T, Samaniego-Castruita D, Singh D, Seumois G, Alzetani A, Woo E, Friedmann PS, King EV, Thomas GJ, Sanchez-Elsner T, Vijayanand P, Ottensmeier CH, Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer, Nat Immunol, 18 (2017) 940–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Savas P, Virassamy B, Ye C, Salim A, Mintoff CP, Caramia F, Salgado R, Byrne DJ, Teo ZL, Dushyanthen S, Byrne A, Wein L, Luen SJ, Poliness C, Nightingale SS, Skandarajah AS, Gyorki DE, Thornton CM, Beavis PA, Fox SB, Darcy PK, Speed TP, Mackay LK, Neeson PJ, Loi S, Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis, Nature medicine, 24 (2018) 986–993. [DOI] [PubMed] [Google Scholar]
- [169].Djenidi F, Adam J, Goubar A, Durgeau A, Meurice G, de Montpreville V, Validire P, Besse B, Mami-Chouaib F, CD8+CD103+ tumor-infiltrating lymphocytes are tumor-specific tissue-resident memory T cells and a prognostic factor for survival in lung cancer patients, J Immunol, 194 (2015) 3475–3486. [DOI] [PubMed] [Google Scholar]
- [170].Pallett LJ, Davies J, Colbeck EJ, Robertson F, Hansi N, Easom NJW, Burton AR, Stegmann KA, Schurich A, Swadling L, Gill US, Male V, Luong T, Gander A, Davidson BR, Kennedy PTF, Maini MK, IL-2(high) tissue-resident T cells in the human liver: Sentinels for hepatotropic infection, J Exp Med, 214 (2017) 1567–1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Zuber J, Shonts B, Lau SP, Obradovic A, Fu J, Yang S, Lambert M, Coley S, Weiner J, Thome J, DeWolf S, Farber DL, Shen Y, Caillat-Zucman S, Bhagat G, Griesemer A, Martinez M, Kato T, Sykes M, Bidirectional intragraft alloreactivity drives the repopulation of human intestinal allografts and correlates with clinical outcome, Sci Immunol, 1 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Snyder ME, Finlayson MO, Connors TJ, Dogra P, Senda T, Bush E, Carpenter D, Marboe C, Benvenuto L, Shah L, Robbins H, Hook JL, Sykes M, D’Ovidio F, Bacchetta M, Sonett JR, Lederer DJ, Arcasoy S, Sims PA, Farber DL, Generation and persistence of human tissue-resident memory T cells in lung transplantation, Sci Immunol, 4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Mackay LK, Minnich M, Kragten NA, Liao Y, Nota B, Seillet C, Zaid A, Man K, Preston S, Freestone D, Braun A, Wynne-Jones E, Behr FM, Stark R, Pellicci DG, Godfrey DI, Belz GT, Pellegrini M, Gebhardt T, Busslinger M, Shi W, Carbone FR, van Lier RA, Kallies A, van Gisbergen KP, Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes, Science (New York, N.Y.), 352 (2016) 459–463. [DOI] [PubMed] [Google Scholar]
- [174].van Gisbergen KP, Kragten NA, Hertoghs KM, Wensveen FM, Jonjic S, Hamann J, Nolte MA, van Lier RA, Mouse Hobit is a homolog of the transcriptional repressor Blimp-1 that regulates NKT cell effector differentiation, Nat Immunol, 13 (2012) 864–871. [DOI] [PubMed] [Google Scholar]
- [175].Rutishauser RL, Martins GA, Kalachikov S, Chandele A, Parish IA, Meffre E, Jacob J, Calame K, Kaech SM, Transcriptional repressor Blimp-1 promotes CD8(+) T cell terminal differentiation and represses the acquisition of central memory T cell properties, Immunity, 31 (2009) 296–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Behr FM, Kragten NAM, Wesselink TH, Nota B, van Lier RAW, Amsen D, Stark R, Hombrink P, van Gisbergen K, Blimp-1 Rather Than Hobit Drives the Formation of Tissue-Resident Memory CD8(+) T Cells in the Lungs, Front Immunol, 10 (2019) 400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Vieira Braga FA, Hertoghs KM, Kragten NA, Doody GM, Barnes NA, Remmerswaal EB, Hsiao CC, Moerland PD, Wouters D, Derks IA, van Stijn A, Demkes M, Hamann J, Eldering E, Nolte MA, Tooze RM, ten Berge IJ, van Gisbergen KP, van Lier RA, Blimp-1 homolog Hobit identifies effector-type lymphocytes in humans, European journal of immunology, 45 (2015) 2945–2958. [DOI] [PubMed] [Google Scholar]
- [178].Cheuk S, Schlums H, Gallais Serezal I, Martini E, Chiang SC, Marquardt N, Gibbs A, Detlofsson E, Introini A, Forkel M, Hoog C, Tjernlund A, Michaelsson J, Folkersen L, Mjosberg J, Blomqvist L, Ehrstrom M, Stahle M, Bryceson YT, Eidsmo L, CD49a Expression Defines Tissue-Resident CD8(+) T Cells Poised for Cytotoxic Function in Human Skin, Immunity, 46 (2017) 287–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Kragten NAM, Behr FM, Vieira Braga FA, Remmerswaal EBM, Wesselink TH, Oja AE, Hombrink P, Kallies A, van Lier RAW, Stark R, van Gisbergen K, Blimp-1 induces and Hobit maintains the cytotoxic mediator granzyme B in CD8 T cells, European journal of immunology, 48 (2018) 1644–1662. [DOI] [PubMed] [Google Scholar]
- [180].Milner JJ, Toma C, Yu B, Zhang K, Omilusik K, Phan AT, Wang D, Getzler AJ, Nguyen T, Crotty S, Wang W, Pipkin ME, Goldrath AW, Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumors, Nature, 552 (2017) 253–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Taniuchi I, Osato M, Egawa T, Sunshine MJ, Bae SC, Komori T, Ito Y, Littman DR, Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development, Cell, 111 (2002) 621–633. [DOI] [PubMed] [Google Scholar]
- [182].Cruz-Guilloty F, Pipkin ME, Djuretic IM, Levanon D, Lotem J, Lichtenheld MG, Groner Y, Rao A, Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs, J Exp Med, 206 (2009) 51–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Shan Q, Zeng Z, Xing S, Li F, Hartwig SM, Gullicksrud JA, Kurup SP, Van Braeckel-Budimir N, Su Y, Martin MD, Varga SM, Taniuchi I, Harty JT, Peng W, Badovinac VP, Xue HH, Runx3 guards cytotoxic CD8+ effector T cells against deviation towards TFH cell lineage, Nat Immunol, 18 (2017) 931–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Hou S, Clement RL, Diallo A, Blazar BR, Rudensky AY, Sharpe AH, Sage PT, FoxP3 and Ezh2 regulate Tfr cell suppressive function and transcriptional program, J Exp Med, 216 (2019) 605–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Paz K, Flynn R, Du J, Qi J, Luznik L, Maillard I, MacDonald KP, Hill GR, Serody JS, Murphy WJ, Sage PT, Sharpe AH, Miklos D, Cutler CS, Koreth J, Antin JH, Soiffer RJ, Ritz J, Bradner JE, Melnick AM, Blazar BR, Small-molecule BCL6 inhibitor effectively treats mice with nonsclerodermatous chronic graft-versus-host disease, Blood, 133 (2019) 94–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Paz K, Flynn R, Du J, Tannheimer S, Johnson AJ, Dong S, Stark AK, Okkenhaug K, Panoskaltsis-Mortari A, Sage PT, Sharpe AH, Luznik L, Ritz J, Soiffer RJ, Cutler CS, Koreth J, Antin JH, Miklos DB, MacDonald KP, Hill GR, Maillard I, Serody JS, Murphy WJ, Munn DH, Feser C, Zaiken M, Vanhaesebroeck B, Turka LA, Byrd JC, Blazar BR, Targeting PI3Kdelta function for amelioration of murine chronic graft-versus-host disease, Am J Transplant, 19 (2019) 1820–1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Badell IR, La Muraglia GM 2nd, Liu D, Wagener ME, Ding G, Ford ML, Selective CD28 Blockade Results in Superior Inhibition of Donor-Specific T Follicular Helper Cell and Antibody Responses Relative to CTLA4-Ig, Am J Transplant, 18 (2018) 89–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Chen Y, Yu M, Zheng Y, Fu G, Xin G, Zhu W, Luo L, Burns R, Li QZ, Dent AL, Zhu N, Cui W, Malherbe L, Wen R, Wang D, CXCR5(+)PD-1(+) follicular helper CD8 T cells control B cell tolerance, Nat Commun, 10 (2019) 4415. [DOI] [PMC free article] [PubMed] [Google Scholar]