Next Article in Journal
Glutamate–Transporter Unbinding in Probabilistic Synaptic Environment Facilitates Activation of Distant NMDA Receptors
Previous Article in Journal
A Systematic Review on Quiescent State Research Approaches in S. cerevisiae
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

PD-L1’s Role in Preventing Alloreactive T Cell Responses Following Hematopoietic and Organ Transplant

BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
*
Author to whom correspondence should be addressed.
Cells 2023, 12(12), 1609; https://doi.org/10.3390/cells12121609
Submission received: 2 May 2023 / Revised: 5 June 2023 / Accepted: 6 June 2023 / Published: 12 June 2023
(This article belongs to the Collection Emerging Use of Stem Cells in Personalized Medicine)

Abstract

:
Over the past decade, Programmed Death-Ligand 1 (PD-L1) has emerged as a prominent target for cancer immunotherapies. However, its potential as an immunosuppressive therapy has been limited. In this review, we present the immunological basis of graft rejection and graft-versus-host disease (GVHD), followed by a summary of biologically relevant molecular interactions of both PD-L1 and Programmed Cell Death Protein 1 (PD-1). Finally, we present a translational perspective on how PD-L1 can interrupt alloreactive-driven processes to increase immune tolerance. Unlike most current therapies that block PD-L1 and/or its interaction with PD-1, this review focuses on how upregulation or reversed sequestration of this ligand may reduce autoimmunity, ameliorate GVHD, and enhance graft survival following organ transplant.

1. Introduction

Self-tolerance is developed by T cells in the thymus. Thymocytes with T cell receptors (TCR) that have a strong affinity for self-peptides presented by the major histocompatibility complex (MHC), or the MHC complex itself, undergo negative selection, resulting in self-reactive thymocytes that undergo apoptosis and do not mature into T cells. However, in organ transplant cases where the tissue within a host is a foreign entity, alloreactivity can develop as central tolerance does not develop towards the non-self/allogenic peptides [1,2]. This occurs in the case of chronic graft rejection, as portions of donor HLA molecules are presented by antigen-presenting cells (APC) in the same manner as foreign bacterial antigens. In acute rejection, TCRs instead identify whole human leukocyte antigen (HLA) molecules presented by the donor cells as foreign. Due to these rejection processes, almost all organ transplants have limited life spans, requiring immunosuppressive therapy in recipients to prolong their graft’s viability [3,4].
HLA matching is crucial for successful hematopoietic stem cell transplantation and to prevent graft-versus-host disease (GVHD). When there is a mismatch between the donor and recipient HLA, donor T cells may mistake host self-peptides for infectious agents due to a process called molecular mimicry. This happens when a donor MHC class I or II molecule displays a common self-peptide that is not normally immunogenic but appears infectious due to variances in the host’s MHC structure. In contrast, HLA-matched donors present alloantigens or minor histocompatibility antigens (mHAs), which can cause GVHD when they are presented by host MHC. mHAs are polymorphic peptides that differ between donor and host cells, and they are positioned within the peptide-binding groove on host MHCs. The binding of mHAs with donor TCRs can lead to an alloreaction where donor T cells attack host tissues [5,6]. GVHD affects up to 80% of mismatched hematopoietic cell transplant (HCT) recipients, and even fully HLA-matched related donors can still result in GVHD in approximately 40% of recipients [7,8].
In both HCT and organ transplantation, alloreactive T cells are activated, often targeting the foreign organ or organs through their presentation of alloantigen [9]. Therefore, a major challenge in these procedures is preserving functional host/graft tissues from alloreactive T cells. This immune response is primarily coordinated by the recognition of MHC and its associated antigen by APCs, which induce subsequent T cell responses. Depending on the subsequent presentation of co-stimulatory and/or co-inhibitory molecules by the APC in association with MHC, co-modulatory molecules can either enhance or weaken an inflammatory response against a target antigen [10]. This article will focus on how activation of T cells by the co-inhibitory molecule PD-L1 has been shown to reduce autoimmunity, symptoms of GVHD, and preserve organ transplants. It is worth noting that PD-L2, which has similar effects as PD-L1, has recently been discovered but will not be discussed here, as PD-L1 appears to be more biologically relevant and has been more widely studied.

2. Current Challenges in Transplantation

2.1. Graft versus Host Disease

One of the most common complications of hematopoietic transplantation therapy is GVHD. This is characterized by transplanted immune cells’ recognition of their new host as foreign and is followed by an immune response against the host following a HCT. GVHD is scored to describe the disease’s severity and functional impact on specific organs. This alloreactive condition can be categorized as either acute or chronic [11,12]
Acute graft versus host disease (aGVHD) is a major cause of morbidity and mortality in HCT. Up to 40% of all cell transplant recipients develop aGVHD [13]. Clinical manifestations of aGVHD most commonly occur within 100 days of transplantation, but acute cases can occur past this time point. Stage 1 aGVHD has limited skin involvement and can present with maculopapular rash, erythema, and pruritus. More severe diseases, stages 2–4, tend to involve the liver and/or gastrointestinal tract. This can present as hyperbilirubinemia, jaundice, and cholestatic hepatitis with liver involvement, as well as severe nausea, vomiting, weight loss, and diarrhea in the gastrointestinal tract [11,12,13,14,15]. However, extensive skin involvement is also possible, presenting as generalized erythroderma that covers greater than 50% of the body’s surface area with additional areas of bullous or desquamation [16].
The pathophysiological mechanism of aGVHD first involves generalized tissue damage induced by radiation or chemotherapeutic agents used as a conditioning regimen before stem cell transplantation [16]. This damage to host tissues releases Damage Associated Molecular Patterns (DAMPs), stimulating the release of a variety of inflammatory cytokines such as IL-1, IL-6, and TNF-α from host non-hematopoietic and remaining host hematopoietic cells. These cytokines upregulate MHC presentation and cell surface adhesion molecules on host APCs, priming the host’s organs for targeting by a subset of T cells that are termed “alloreactive” [17,18]. Focusing on the GI system, destruction of rapidly replicating epithelial cells in the intestine results in increased permeability of the mucosa, followed by the release of microbial components such as LPS and other pathogen-associated molecular patterns (PAMPs) into systemic circulation. Host DCs are then activated by these PAMPs and the aforementioned DAMPs. Recognition of these DAMPs and PAMPs by toll-like receptors causes these DCs to begin increasing the expression of alloantigen and CCR7, which allow for their migration into mesenteric lymph nodes. The upregulation of MHC, surface adhesion molecules, and migration of alloreactive DCs primes the transplant recipient for the initial onset of aGVHD [19,20,21].
Host and new donor APCs then activate alloreactive donor T cells still present in the hematopoietic transplant that aggregate in secondary lymphoid tissue [22,23]. This results in increased proliferation, trafficking, and activation of both CD4+ and CD8+ T cells, with T cell activation further inducing the release of IL-2 and IFN-γ [24,25,26,27,28]. Finally, alloreactive effector T cells (including helper T cells and cytotoxic T cells), along with natural killer (NK) cells, migrate to various organ systems where they begin to cause cytotoxic tissue damage. This eventually leads to the characteristic manifestations of aGVHD [29,30,31].
In contrast to aGVHD, chronic graft versus host disease (cGVHD) typically occurs 100 or more days after transplantation, affecting 30–70% of transplant recipients [32,33,34]. Clinical manifestations of chronic GVHD can appear in a variety of organ systems and can either be restricted to a single organ or widespread throughout the body. Dermatologic manifestations of chronic GVHD include lichen-sclerosis-like lesions, depigmentation, sweat impairment, and heat intolerance, along with the typical erythema and maculopapular rash. Gastrointestinal features of chronic GVHD include exocrine pancreatic insufficiency, webs, strictures, and stenosis within the esophagus. Pulmonary manifestations include bronchiolitis obliterans and bronchiectasis. Patients with chronic GVHD also experience dry and painful eyes, conjunctivitis, and punctate keratitis [11,12].
The pathophysiological mechanism of cGVHD has been suggested to follow a similar sequence, which culminates in the development of aGVHD [35]. Initially, host tissues are damaged by the transplant conditioning regimen, infectious agents, and previous aGVHD. This damage causes the release of various cytokines and microbial contents, which activate APCs [20,36,37,38,39]. The activation of professional APCs, such as macrophages and DCs, then leads to the subsequent differentiation of donor CD4+ T cells into Th1 and Th17 cells, which are then recruited into host tissues [40,41,42,43]. These effector T cells then infiltrate and cause widespread dysregulated inflammation in various organs, which further feeds into a cycle of inflammation and cell damage. In particular, the thymus and other secondary lymphoid organs are damaged, resulting in the loss of central and peripheral tolerance and the production of autoreactive immune cells [44,45,46,47,48,49,50].
This widespread tissue damage results in a cycle of perpetual inflammation [51,52,53,54,55,56,57,58,59,60,61,62,63,64]. Following the release of cytokines from damaged host tissues, activated immune cells, including APCs, T cells, B cells, and NK cells, continue damaging their host, which in turn perpetuates the release of cytokines. Regulatory T cells (Tregs) that normally act as a break to this inflammatory cycle are nonfunctional following the loss of central and peripheral tolerance [51,52,53,54,55,56,57]. Finally, once tissues have been eviscerated by alloreactive immune cells, there is abnormal tissue repair, which results in the release of TGF-β and widespread tissue fibrosis, ultimately manifesting in the symptoms of cGVHD [58,59,60,61,62,63,64].
aGVHD and cGVHD differ in their pathophysiology and symptomatology, with aGVHD characterized by cytotoxicity and cGHVD by tissue fibrosis. However, central to both processes is T cell activity, meaning an interruption to this T cell alloactivation would protect tissues from the effects of both aGVHD and cGVHD.

2.2. HCT Rejection

Graft failure is a severe complication following HCT that is divided into primary graft failure and secondary graft failure. In primary graft failure, there is a failure to achieve initial engraftment of the HCT. This is defined as an absolute neutrophil count (ANC) lower than 0.5 × 109/L by 28 days, platelet levels less than 20 × 109/L, or hemoglobin levels less than 80 g/L after transplantation. In secondary graft failure, there is a failure of the transplanted cells (ANC lower than 0.5 × 109/L) after initially successful engrafting [65].
The pathogenesis of graft failure in HCT is characterized by a complex, multifactorial, alloreactive immune response from host T cells and NK cells. Graft failure due to host NK cells most commonly occurs following MHC-mismatched HCT. However, even in HLA-matched HCT, host T cells have been shown to promote graft failure [66,67,68]. Host NK cells have inhibitory receptors that are unable to recognize the mismatched MHC class one molecules on the transplanted cells, resulting in NK-mediated cytotoxic rejection of the allogenic cells [69,70]. Host cytotoxic T cells remaining after the conditioning regimen in HCT also play a major role in graft failure. The exact mechanism behind host T cell-mediated graft rejection remains unclear, as the cytotoxic activity of these T cells is independent of typical perforin, Fas-ligand, TNF-related apoptosis-inducing ligand, and TNF Receptor-1 mechanisms [71,72,73,74].
Interestingly, donor NK cells and donor T cells have been shown to promote engraftment by inhibiting the host cell response to the transplanted cells [75,76,77,78]. Both host and donor Treg cells have also been demonstrated to facilitate engraftment in HCT through the production of IL-10 and TGF-β, suppressing T cell and NK cell-mediated graft failure [79,80,81].
Even though graft rejection has an extremely poor prognosis, the condition is relatively rare among allogeneic HCT recipients, with an incidence of roughly 5% [82]. The current treatment for HCT graft failure mainly consists of G-CSF therapy and additional allogenic cells to increase the graft’s proliferative capacity and prevent its rejection [83]. The rarity of HCT graft failure in comparison to rates of GVHD and solid organ rejection makes it no surprise that current research on how PD-L1 functions in HCT rejection is scarce itself.

2.3. The Pathophysiologic Process of Transplant Rejection

Graft failure remains a major limitation in other types of tissue transplantation beyond HCT. Kidney transplant is the most transplanted organ in the United States of America. In a review of United Network for Organ Sharing data on kidney transplantation from 2000 to 2014, 50,301 graft failures occurred, or about 3600 every year. Of this, 48% were due to chronic rejection, and 12% were due to acute rejection [84]. The immunological mechanism of transplant rejection occurs in three main forms: hyperacute, acute, and chronic. Hyperacute rejection (HAR) typically occurs within minutes to hours. Pre-formed circulating antibodies quickly bind to transplant tissue, typically resulting in irreversible graft thrombosis and ischemia. However, with the advent of screening technologies such as crossmatch testing and antibody screening over the last 40 years, HAR is now a rare occurrence [85]. Hyperacute rejection (HAR) is mainly a humoral immune response and is classified as a Type II hypersensitivity reaction. It occurs because pre-formed circulatory antibodies bind to the allogenic graft tissue. These antibodies arise because of previous exposure to foreign immune molecules, typically the constitutively expressed human leukocyte antigen Class I molecules. As such, HAR most frequently occurs in patients who have had previous transplantation, blood transfusion, or pregnancy [86].
In contrast, acute rejection (AR) occurs within days to weeks of transplantation. It is the result of the host immune system identifying the graft as foreign and destroying it, leading to graft failure. This type of rejection is largely controlled by immunosuppressive therapy, leading to a decreased incidence and better long-term outcomes. However, even with treatment, AR still occurs in about 7% of renal transplants every year [87]. Chronic rejection (CR) occurs months to years after transplantation and affects 100% of transplants to some degree. It is one of the main determinants of the longevity of solid organ transplants [88]. Its full etiology has yet to be understood, and proposed pathological mechanisms cite inflammatory, humoral, and cellular immune reactions.
Acute transplant rejection differs from HAR in that it involves both a humoral and cellular immune response. Additionally, in AR, the patient does not have pre-formed antibodies, but rather develops an activated immune response toward the graft tissue. This immune response arises because resident-donor antigen-presenting cells in the graft travel to sites of immune activation in the host, such as lymph nodes, and are recognized by a subset of T cells that are alloreactive. Alloreactive T cells are defined by the ability of their T cell receptors (TCRs) to bind non-self MHC molecules [89]. The mechanism of AR is the direct pathway of allorecognition, in that graft MHC molecules are directly presented to and recognized by host lymphocytes. Activation of alloreactive T cells leads to the typical host immune response to a foreign substance. This includes the activation of CD8+ T cells, Th1 T cells, Th2 T cells, and Follicular Helper T (TFH) cells. CD8+ T cells will migrate to the transplanted tissue and exert their direct cytotoxic effects. Activated Th1 cells will secrete IFN-γ resulting in the activation of macrophages and inflammation. TFH T cells serve as a link to humoral immunity, aiding B cell activation and the generation of donor-specific antibodies (DSAs) [90].
Whereas acute transplant rejection occurs via direct allorecognition, chronic transplant rejection occurs via the indirect pathway of allorecognition. The indirect pathway of allorecognition activates when graft T cells die in a manner that results in the release of cellular alloantigens, such as immune-mediated death or inflammasome-mediated pyroptosis. These cellular alloantigens, including peptide fragments of donor MHC molecules, are then phagocytosed by host APCs. Activated host immune cells travel to sites of immune activation and stimulate an immune response against the transplanted tissue, like the direct pathway of allorecognition [9]. While CR initially follows a similar cellular immune response activation as AR, there are several additional pathological changes that make CR a more complex and severe disease. One of the most distinctive features of chronic solid organ rejection is the activation of vascular smooth muscle cells and fibroblasts, resulting in intimal hyperplasia of blood vessels, and leading to ischemia of graft tissue. While the exact pathogenesis of this is not understood, most hypotheses start with vascular endothelial inflammation due to T cell-mediated release of cytokines. It has been suggested that this immune response can lead to aberrant migration and proliferation of vascular smooth muscle cells [88].
While the exact immunological mechanisms of AR and CR processes are varied, they both share an underlying framework: cellular damage to graft tissue that initiates an immune response driven by T cells. If these processes can be interrupted by limiting immune cell activation, it may promote longevity in engrafted tissues.

3. PD-L1 Mechanism of Action

PD-L1 (also known as B7-H1) and its corresponding receptor, PD-1, are both transmembrane proteins that are a part of the immunoglobulin superfamily. PD-L1 is constitutively present on the surface of hematopoietic cells such as monocytes and T cells. However, monocytes in peripheral blood do not tend to express PD-L1. Certain immune-privileged sites, such as the placenta and cornea, have also been shown to maintain expression of PD-L1. Additionally, the ligand’s expression is upregulated during active immune responses. This upregulation has been linked to the presence of pro-inflammatory cytokines such as IFN-γ or exogenous LPS on the surface of hematopoietic cells and at the surface of their epithelial and endothelial cells [28,91,92]. PD-L1 mRNA has been found in human organs, including the heart, kidney, and lung, but not in the colon or small intestine [93]. In contrast to this finding, immunohistochemistry stains performed in mice differ from the mRNA expression profiles seen in humans. Stains still show PD-L1 protein expression in the endothelium of the heart and macrophages in the lung. However, in contrast to the mRNA expression profiles of humans, mice express PD-L1 proteins in the lamina propria of the small intestine but not in the kidneys [92].
Although soluble versions of PD-L1 and PD-1 exist, conventionally, the ligand and its receptor are transmembrane-bound proteins that are displayed on the cell’s surface. More specifically, PD-1 has an extracellular domain that allows for interaction with PD-L1, a localizing transmembrane portion, and an intracellular domain that allows for signal transduction [94]. The receptor’s production is upregulated via TCR recognition of MHC, thereby preventing the overactivation of T cells and limiting immune-mediated damage to native tissue [95,96]. PD-L1 has been shown to decrease the expansion of CD4+ T cells and NK cells, limit the cytotoxic effects of CD8+ T cells, and simultaneously increase the differentiation of CD4+ T cells into Tregs [97,98,99]. PD-1 can be stimulated following TCR activation in a two-step sequence whereby TCR engagement with MHC is followed by co-inhibition by PD-L1 [100]. Once PD-L1 is recognized by PD-1 in peripheral (non-immune) tissues, it initiates a co-inhibitory signal, limiting T cell proliferation (Figure 1b). This immunomodulatory effect is driven by the downregulation of a variety of intracellular pathways responsible for cellular metabolism and proliferation.
Crucial to this intracellular regulation are intracellular signaling domains on the PD-1 molecule known as immunoreceptor tyrosine-based switch motifs (ITSM). Following TRC activation and PD-1/PD-L1 binding, these ITSMs are phosphorylated, allowing for their interaction with a cytoplasmic tyrosine phosphatase called Src homology 2 domain-containing phosphatase 2 (SHP-2). SHP-2 contains an enzymatic tail portion that allows for downstream inhibition of phosphatidylinositol-3-kinase (PI3K) and Akt as well as a reduction in IL-2 production [101,102,103,104]. PD-L1’s inhibition of PI3K/AKT thereby directly counteracts the effects exerted by CD80 (also known as B7-1), a costimulatory molecule present on APCs [105,106] (Figure 1a). This prevents T cell activation via two alternate mechanisms: the loss of IL-2, preventing T cell expansion, and the limitation of the PI3K/AkT cell-signaling cascade, reducing glucose metabolism. Thereby, CD8+ and CD4+ T cells are prevented from proliferating and exerting their effector functions. This cumulative effect allows PD-L1 to act as a break on lymphocyte activity, preventing an overactive immune response [107,108,109].
There is an additional interaction between PD-L1 and another B7 immunoregulatory ligand, CD80. This interaction has not been as extensively studied as the PD-L1/PD-1 pathway, so the mechanism of the PD-L1/CD80 interaction is not entirely clear. Reports on the PD-L1 binding of CD80 have differed, but recent data indicates that their interactions are restricted to cis-membrane interactions and that the molecules do not interact trans-cellularly. This means that CD80 and PD-L1 are restricted to interactions on the same cellular membrane as they combine to form a heterodimer [110,111].
The CD80/PD-L1 interaction on the surface of APCs is pro-inflammatory, as CD80 inhibits PD-L1’s binding of PD-1, while CD80’s capacity to bind CD28 is preserved. That is to say, in the presence of both equal portions of CD80 and PD-L1, CD80 costimulatory signaling would tend to predominate on the surface of professional APCs [110,111,112]. However, the PD-L1/CD80 interaction also seems to be involved with the promotion of Treg expansion and T cell tolerance [113,114]. Additionally, the expression of CD80 by CD8+ T cells prevents this population of cells from proliferating [115]. Taken together, these findings support the theory positing that the effect of the PD-L1/CD80 interaction is dependent on the cell type that expresses the two proteins. Although the interaction between PD-L1 and CD80 offers PD-L1 an additional pathway to regulate T cell survival, this is a relatively new discovery, and the exact mechanisms underlying these molecules’ interaction have yet to be elucidated [110].

4. PD-L1 Amelioration of GVHD, Autoimmunity, and Graft Rejection

4.1. GVHD

A major goal of therapeutics following hematopoietic transplantation is to prevent the development of GVHD. PD-L1 appears to have the potential to promote immune tolerance following transplant and minimize the potential for developing adverse reactions caused by stem cell therapy. Tang et al. demonstrated how in vivo hydrodynamic gene transfer, which induces overexpression of PD-L1 in murine aGVHD models, results in lower lethality due to aGVHD. This overexpression of PD-L1 was found to result in the inhibition of donor T cells and reduce effector memory status. Moreover, PD-L1 overexpression resulted in murine models exhibiting lower levels of pro-inflammatory cytokines secreted from effector T cells, less proliferation, and increases in the apoptotic activity of these effector T cells [116].
A similar experiment performed by Blazar et al. found that blocking the PD-1 receptor from engaging with PD-L1 accelerated the lethality of GVHD in murine models. Following GVHD induction using donor splenocytes, anti-murine PD-1 monoclonal antibodies (mAB) or PD-L1 fusion proteins were administered, which block PD-1 signaling with its ligand. The PD-1 blockade was found to enhance T cell alloresponsiveness both in vitro and in vivo. This alloresponse was associated with a significant increase in inflammatory cytokines such as TNF-α and IFN-γ and was thought to cause a significant acceleration in GVHD lethality. Notably, analysis of the murine liver tissue revealed that irrelevant mAB (meaning non-PD-1-blocking)-treated recipient mice had less severe injuries when compared to those given anti-PD-1 mAB [117].
It is important to note that the development of GVHD is dependent on which cells express PD-L1. While expression of the ligand on donor T cells induces GVHD, expression on parenchymal tissue such as hepatocytes results in protection from the development of GVHD [118,119].
Deng et al. further characterized the PD-L1/CD80 interaction by administering anti-PD-L1 monoclonal antibodies. These antibodies were designed to block PD-L1’s interaction with CD80 without interfering with the PD-L1/PD-1 pathway. Administration of the antibody was performed on mice with intact PD-1 and rat-IgG in a control group for five days following a HCT [114]. After inducing GVHD via injection of donor spleen cells, Deng et al. found that in mouse models with intact PD-1, administration of these blocking antibodies reduced proliferation and apoptosis of donor alloreactive T cells. Overall, this resulted in a relative reduction in proliferation that was less marked than the reduction in apoptosis and allowed for an expansion of alloreactive donor T cells, producing more severe GVHD. The experimental group showed 100% mortality by day 7, following the administration of the PD-L1 antibody, while 50% of the control group survived for >50 days (N = 10). Cellular analysis showed that there was a marked increase in CD4+ donor T cells in the experimental group, as well as a decrease in expression of PD-1, quantified by RT-PCR. Cytokine analysis following blockage of PD-L1/CD80 showed that there was a significant decrease in IL-2 production as well as an increase in IFN-γ and TNF-α found in the serum. The increase in IFN-γ and TNF-α was attributed to increased numbers of alloreactive T cell populations in the experimental cohort. Additionally, the decreased IL-2 concentration was associated with decreased PD-1 production by both CD4+ and CD8+ T cells [120]. Intracellularly, the block on PD-L1/CD80 interactions increased the expression of BCL-xL, a pro-survival gene, and reduced the expression of caspase-3 in donor T cells. Notably, the ameliorative effect that the PD-L1/CD80 interaction has on GVHD depends on the presence of PD-1 interactions. Blocking the PD-L1/CD80 interaction in PD-1−/− mice instead alleviates GVHD when compared to PD-1−/− mice that had not received the blocking antibody. Overall, the results from this experiment demonstrated that the PD-L1/CD80 interaction prevents GVHD in a PD-1-dependent manner [111].
Deng et al. also found that injection with PD-L1 fused to an Ig Fc domain led to an improvement in GVHD symptoms in mice with intact PD-1. This is because there is a crucial balance between proliferation and apoptosis in alloreactive T cells. In a PD-L1−/− cohort of mice, it was found that following injection of a PD-L1 Ig-expressing plasmid, there was a significant reduction of donor CD4+ T cells in the spleen and liver of the experimental group, resulting in less severe GVHD. In comparison, the control group without the PD-L1 plasmid injection developed severe acute GVHD and died within 7 days of the transplant. Interestingly, this experiment was repeated with a PD-1−/− and control group model that contrasted these results. In PD-1−/− mice, injection of a PD-L1 plasmid led to rapid loss of body weight and death 15 days post-HCT. Overall, there was an increase in the severity of GVHD, as demonstrated by the increase in mortality, symptomatology, and expansion of alloreactive donor CD4+ T cells. The results of Deng et al. reinforce the notion that the interaction between PD-L1 and CD80 leads to augmented proliferation of activated T cells and that PD-L1’s interaction with the PD-1 receptor causes a crucial increase in alloreactive T cell apoptosis. This balance is paramount to survival post-transplant [111].

4.2. Autoimmunity

The mechanism by which PD-L1 plays a role in autoimmune disease can be translated thematically to the expression of PD-L1 during cell transplantation. A study by Hu et al., using DBA/1J mouse models (a strain of mice bred to develop rheumatoid arthritis), studied the impact of PD-L1 overexpression on these mice. Researchers administered a PD-L1 lentiviral vector to transfect mouse bone marrow mesenchymal cells, which induced PD-L1 overexpression in the cells. DBA/1J mice that were treated with mesenchymal stem cells that had received the PD-L1 vector exhibited less joint damage, less activation of pro-inflammatory cytokines, and an inhibition of T and B cell activation. Moreover, there was limited cartilage damage, synovial hyperplasia, inflammatory cell infiltration, and bone erosion in the mice treated with the PD-L1 overexpression vector. These transplanted stem cells decreased the DCs found in mouse joints and increased the Tregs in the tissue, promoting a tolerant and non-inflammatory environment [121].
In addition to the PD-L1/PD-1 interaction, the PD-L1/CD80 interaction is emerging as a significant pathway in terms of regulating autoimmunity. Sugiura et al. created an antibody that binds to CD80, disrupting the PD-L1/CD80 interaction while mostly sparing CD80’s interaction with both CD28 and CTLA-4. Administration of the novel CD80 antibody to mice with model autoimmune conditions, including rheumatoid arthritis and autoimmune encephalomyelitis, showed significant improvement in clinical and histologic scores of disease severity. Additionally, administration of the PD-L1/CD80 interaction blocking antibody decreased inflammatory markers such as IL-17 and IFN-γ as well as significantly decreased lymphocyte infiltration into glandular tissue in the case of mouse-modeled Sjogren syndrome [122].
The PD-L1/PD-1 interaction is crucial in the regulation of lymphocyte responses in various other autoimmune diseases. PD-L1’s absence has been shown to exacerbate a variety of immune-mediated disease processes such as encephalomyelitis, lupus-like nephritis, and autoimmune dilated cardiomyopathy in mouse models [123,124,125]. This shows that there is potential for using PD-L1 overexpression to decrease immune cell function and subsequent pathologies that may be associated with heightened immune cell function following organ transplant and in autoimmune conditions.

4.3. Solid Organ Tolerance

In addition to PD-L1’s role in GVHD prevention, the role this ligand holds in organ transplantation has also been explored. Tanaka et al. studied the effects of PD-L1’s presence in mice that received a mismatched cardiac allograft that underwent concurrent immunosuppression with CTLA4Ig. Their study showed that blocking PD-L1 resulted in significantly reduced numbers of Tregs and significantly increased portions of alloreactive cytotoxic and IFN-γ-producing T cells within the spleen of allograft recipient mice. Regarding allograft function, Tanaka et al. demonstrated that PD-L1 blocking antibodies induced graft rejection within this same population of transplanted mice (Figure 2a,b). Moreover, PD-L1-deficient mice receiving heart transplants also had a significant reduction in graft survival. Histologically, it seems a possible cause of these effects in PD-L1 knockout mice was the increased vasculopathy within the cardiac tissue of mice [126].
Another study that focused on liver transplants observed how knockout of PD-1 receptors on murine T cells results in rapid liver allograft rejection in comparison to wild-type mice. Similarly, when treated with anti-PD-L1 antibodies, allograft recipient mice had significantly reduced survival rates as compared to allograft recipients not receiving anti-PD-L1 antibodies. Upon closer examination, allograft recipients who were treated with anti-PD-L1 antibodies revealed histologic liver sections that had significant parenchymal necrosis and vasculature inflammation, as well as drastic elevations in inflammatory markers like perforin/granzyme and iNOS mRNA within the grafted tissue [127].
An observational study of pediatric liver transplant patients observed an elevated ratio of PD-L1 to CD86 (a costimulatory molecule similar in function to CD80) within a population of DCs. There was a significant association between these patients’ diminished need for immunosuppressive therapy following transplant. In this same subset of patients, a relatively higher expression of PD-L1 on DCs was also directly correlated with higher levels of Tregs. However, it should be noted that this study did not show a correlation between a low PD-L1:CD86 ratio and low levels of Tregs [128]. Together, these studies reveal that PD-L1 may serve to protect allograft function by preventing immune-mediated tissue damage and promoting graft survival. However, further studies are needed to definitively prove or dispute PD-L1’s role in solid organ transplantation.
Figure 2. Proposed protective effects of PD-L1 during transplantation. (a) Limited PD-L1 expression by transplanted cells provides less protection from cytotoxic T cell infiltration (and less protection from helper T cell infiltration). Inflammation progresses to acute rejection and eventual graft failure as the tissue loses viable graft cells. (b) PD-L1 may be upregulated by methods such as IFN-γ administration or hydrodynamic gene transfer onto transplanted DCs and certain graft tissues such as hepatocytes [93,116,118,129]. If PD-L1 upregulation is sufficient to overcome CD80 sequestration/signaling, a reduction in alloreactive T cells would prevent graft cell induced apoptosis. Ideally, this technique would preserve graft function by preventing T cell infiltration.
Figure 2. Proposed protective effects of PD-L1 during transplantation. (a) Limited PD-L1 expression by transplanted cells provides less protection from cytotoxic T cell infiltration (and less protection from helper T cell infiltration). Inflammation progresses to acute rejection and eventual graft failure as the tissue loses viable graft cells. (b) PD-L1 may be upregulated by methods such as IFN-γ administration or hydrodynamic gene transfer onto transplanted DCs and certain graft tissues such as hepatocytes [93,116,118,129]. If PD-L1 upregulation is sufficient to overcome CD80 sequestration/signaling, a reduction in alloreactive T cells would prevent graft cell induced apoptosis. Ideally, this technique would preserve graft function by preventing T cell infiltration.
Cells 12 01609 g002

4.4. PD-L1 Viral Reactivation

The potential of PD-L1 to prevent transplant rejection and graft survival must be weighed against its immunosuppressive properties, especially in the case of viral reactivation. The expression of PD-L1 on CD8+ T cells decreases the proliferative capacity and cytotoxic function of these cells. Given these effects, it is not surprising that many viruses induce PD-L1 overexpression, thereby decreasing the host cytotoxic T cell response and contributing to viral immune escape. Recently implicated viruses include Hepatitis B virus (HBV), HIV, Epstein–Barr virus, hantaviruses, and even coronaviruses [130,131,132,133,134]. Consequently, any therapy involving the upregulation of PD-L1 represents a clear pathway for the reactivation of underlying viral infections.
However, the effect of any immunotherapy interacting with PD-L1 is not so simple. While one would expect that any immunotherapy downregulating PD-L1 would lead to increased immune function and, therefore, decrease viral reactivations, there is clinical evidence of increased rates of HBV reactivation in cancer patients being treated with anti-PD-L1 monoclonal antibodies [135]. The exact mechanism by which this occurs is still unclear. Potential theories include the role of PD-1 in preventing immune-associated hepatocyte damage or the role of PD-1 in Treg apoptosis [43,136,137]. Overall, there is much to be discovered about the effects of PD-L1 immunotherapies in vivo.
Additionally, the potential adverse effects of PD-L1-induced immunosuppression must be viewed in the context of the current gold standard of post-transplant and GVHD immunosuppressive therapies, such as calcineurin inhibitors and corticosteroids. The high rates of infection in post-transplant patients due to immunosuppression are already well understood [4,138]. This is especially evident in the occurrence of viral reactivation. Traditional immunosuppressive therapies after solid organ transplant have been strongly implicated in the reactivation of Epstein–Barr virus, HBV, BK polyomavirus, and many more [138,139,140,141]. Overall, the potential immunosuppressive effects of PD-L1 must be further investigated, as it represents a promising alternative to current traditional immunosuppressive therapies.

5. Current State of PD-L1/PD-1 Targeting Therapies

Pharmaceutical targeting of PD-L1 has primarily focused on the inhibition of excess PD-L1 produced by cancer cells. Several monoclonal antibodies have been approved to target the PD-L1/PD-1 interaction, including pembrolizumab, nivolumab, and cemiplibab. Atezolizumab, avelumab, and durvalumab are all approved to target and inhibit PD-L1 [130]. Each of these therapeutics seeks to inhibit the immunosuppression seen in tumor cells that overexpress PD-L1, preventing immune tolerance in cancerous growth. However, these treatments have also been found to lead to the development of self-reactive T cells and have been linked to the development of autoimmune and inflammatory diseases [131].
In the absence of PD-L1/PD-1-induced anergy, patients receiving treatment that inhibits this pathway are at increased risk for developing inflammatory arthritis as well as hematologic diseases, including immune thrombocytopenia and autoimmune hemolytic anemia [132,133]. To circumvent some of the adverse effects noted in patients undergoing treatment with monoclonal antibodies, some researchers are pursuing combination therapies with small molecules such as osimertinib, an EGFR inhibitor, that mutually function to limit the over-activation of PD-L1. These combination therapies are in development for the treatment of lymphomas, NSCLC, colon cancer, and melanoma [134].
Other researchers have considered targeting non-membranous PD-L1 expression with small molecules that inhibit the expression of PD-L1 in its nuclear, cytoplasmic, or extra-cellular vesicle expression to ultimately reduce signaling sequelae [135,136]. Another avenue for investigation into PD-L1/PD-1 inhibition is more indirect by targeting its expression via known regulators of the PD-L1/PD-1 pathway, such as epigenetic modification and transcriptional and post-transcriptional regulators [137]. These emerging immunotherapies offer a variety of methods to enhance the body’s ability to target and kill cancer cells in a more precise manner. Researchers’ focus on PD-L1 and its role in the protection of cancer through anergy has resulted in prolonged lives for many. However, up until this point in time, researchers’ goal with PD-L1 targeting therapy has been to increase immune response via inhibition of the ligand. Therapeutic induction of PD-L1 expression seeks the opposite goal from most current pharmacologic agents in development—to promote local immunosuppression.

6. Conclusions

One major limitation shared by hematopoietic cellular therapies and solid organ transplantation is the occurrence of graft-versus-host disease (GVHD) and graft failure, respectively. In both cases, an overactive immune system targets and destroys functional tissue, similar to autoimmune diseases.
Promising targets for future immunotherapy include the interaction of PD-L1 with PD-1 or CD-80, which play a role in regulating T cells. Studies have shown that overexpression of PD-L1 in GVHD models improves survival and reduces pro-inflammatory cytokine secretion. Similarly, stimulating PD-L1 during organ transplantation prolongs the graft’s lifespan and reduces rejection rates. Additionally, in autoimmune disease models, increased PD-L1 expression leads to slower disease progression and limited tissue damage.
Although PD-L1 has primarily been studied in the context of cancer treatment, it has the potential to be a valuable tool in the management of allo- and auto-immunity. While most research has focused on blocking PD-L1 to inhibit immunosuppressive processes, enhancing PD-L1 activity could be a promising therapeutic approach as it dampens a destructive immune reaction. Further investigation is needed to understand how increased PD-L1 expression and availability in the context of GVHD, autoimmunity, and organ transplantation can promote immune tolerance.

Author Contributions

Conceptualization, Y.L., J.O. and S.H.; investigation, S.H., K.C., J.L., D.H. and J.O.; resources, Y.L.; data curation, S.H., K.C., J.L., D.H. and J.O.; writing—original draft preparation, S.H., K.C., J.L., D.H. and J.O.; writing—review and editing, S.H. and J.O.; visualization, S.H.; supervision, Y.L.; project administration, Y.L. and S.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Biorender was used to create Figure 1 and Figure 2.

Conflicts of Interest

Yung Li is one of the executive editors of the Journal of Cellular Biochemistry. All other authors declare no conflict of interest.

References

  1. Xing, Y.; Hogquist, K.A. T-cell tolerance: Central and peripheral. Cold Spring Harb. Perspect. Biol. 2012, 4, a006957. [Google Scholar] [CrossRef] [Green Version]
  2. Felix, N.J.; Allen, P.M. Specificity of T-cell alloreactivity. Nat. Rev. Immunol. 2007, 7, 942–953. [Google Scholar] [CrossRef]
  3. García, M.A.A.; Yebra, B.G.; Flores, A.L.L.; Guerra, E.G. The major histocompatibility complex in transplantation. J. Transpl. 2012, 2012, 842141. [Google Scholar] [CrossRef]
  4. Nelson, J.; Alvey, N.; Bowman, L.; Schulte, J.; Segovia, M.C.; McDermott, J.; Te, H.S.; Kapila, N.; Levine, D.J.; Gottlieb, R.L.; et al. Consensus recommendations for use of maintenance immunosuppression in solid organ transplantation: Endorsed by the American College of Clinical Pharmacy, American Society of Transplantation, and International Society for Heart and Lung Transplantation: An executive summary. Pharmacotherapy 2022, 42, 594–598. [Google Scholar] [CrossRef]
  5. Spierings, E. Minor histocompatibility antigens: Past, present, and future. Tissue Antigens 2014, 84, 360. [Google Scholar] [CrossRef] [PubMed]
  6. Koyama, M.; Hill, G.R. Alloantigen presentation and graft-versus-host disease: Fuel for the fire. Blood 2016, 127, 2963–2970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Ferrara, J.L.M.; Levine, J.E.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef] [PubMed]
  8. Spellman, S.R. Hematology 2022-what is complete HLA match in 2022? Hematol. Am. Soc. Hematol. Educ. Program 2022, 2022, 83–89. [Google Scholar] [CrossRef]
  9. Siu, J.H.Y.; Surendrakumar, V.; Richards, J.A.; Pettigrew, G.J. T cell Allorecognition Pathways in Solid Organ Transplantation. Front. Immunol. 2018, 9, 2548. [Google Scholar] [CrossRef] [Green Version]
  10. Sharpe, A.H. Mechanisms of costimulation. Immunol. Rev. 2009, 229, 5–11. [Google Scholar] [CrossRef] [Green Version]
  11. Filipovich, A.H.; Weisdorf, D.; Pavletic, S.; Socie, G.; Wingard, J.R.; Lee, S.J.; Martin, P.; Chien, J.; Przepiorka, D.; Couriel, D.; et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol. Blood Marrow Transpl. 2005, 11, 945–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Jagasia, M.H.; Greinix, H.T.; Arora, M.; Williams, K.M.; Wolff, D.; Cowen, E.W.; Palmer, J.; Weisdorf, D.; Treister, N.S.; Cheng, G.-S.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol. Blood Marrow Transpl. 2015, 21, 389–401.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Weisdorf, D.; Haake, R.; Blazar, B.; Miller, W.; McGlave, P.; Ramsay, N.; Kersey, J.; Filipovich, A. Treatment of moderate/severe acute graft-versus-host disease after allogeneic bone marrow transplantation: An analysis of clinical risk features and outcome. Blood 1990, 75, 1024–1030. [Google Scholar] [CrossRef] [Green Version]
  14. Harris, A.C.; Young, R.; Devine, S.; Hogan, W.J.; Ayuk, F.; Bunworasate, U.; Chanswangphuwana, C.; Efebera, Y.A.; Holler, E.; Litzow, M.; et al. International, Multicenter Standardization of Acute Graft-versus-Host Disease Clinical Data Collection: A Report from the Mount Sinai Acute GVHD International Consortium. Biol. Blood Marrow Transpl. 2016, 22, 4–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Zeiser, R.; Blazar, B.R. Acute Graft-versus-Host Disease—Biologic Process, Prevention, and Therapy. N. Engl. J. Med. 2017, 377, 2167–2179. [Google Scholar] [CrossRef]
  16. Flinn, A.M.; Gennery, A.R. Treatment of Pediatric Acute Graft-versus-Host Disease-Lessons from Primary Immunodeficiency? Front. Immunol. 2017, 8, 328. [Google Scholar] [CrossRef] [Green Version]
  17. Alegre, M.L.; Lakkis, F.G.; Morelli, A.E. Antigen Presentation in Transplantation. Trends Immunol. 2016, 37, 831–843. [Google Scholar] [CrossRef]
  18. Hill, G.R.; Koyama, M. Cytokines and costimulation in acute graft-versus-host disease. Blood 2020, 136, 418–428. [Google Scholar] [CrossRef]
  19. Xun, C.Q.; Thompson, J.S.; Jennings, C.D.; Brown, S.A.; Widmer, M.B. Effect of total body irradiation, busulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-versus-host disease in H-2-incompatible transplanted SCID mice. Blood 1994, 83, 2360–2367. [Google Scholar] [CrossRef] [Green Version]
  20. Cooke, K.R.; Gerbitz, A.; Crawford, J.M.; Teshima, T.; Hill, G.R.; Tesolin, A.; Rossignol, D.P.; Ferrara, J.L. LPS antagonism reduces graft-versus-host disease and preserves graft-versus-leukemia activity after experimental bone marrow transplantation. J. Clin. Investig. 2001, 107, 1581–1589. [Google Scholar] [CrossRef] [Green Version]
  21. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev 2009, 22, 240–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Duffner, U.A.; Maeda, Y.; Cooke, K.R.; Reddy, P.; Ordemann, R.; Liu, C.; Ferrara, J.L.M.; Teshima, T. Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease. J. Immunol. 2004, 172, 7393–7398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gao, E.K.; Kosaka, H.; Surh, C.D.; Sprent, J.T. Cell contact with Ia antigens on nonhemopoietic cells in vivo can lead to immunity rather than tolerance. J. Exp. Med. 1991, 174, 435–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Chakraverty, R.; Sykes, M. The role of antigen-presenting cells in triggering graft-versus-host disease and graft-versus-leukemia. Blood 2007, 110, 9–17. [Google Scholar] [CrossRef]
  25. Mohty, M.; Blaise, D.; Faucher, C.; Vey, N.; Bouabdallah, R.; Stoppa, A.-M.; Viret, F.; Gravis, G.; Olive, D.; Gaugler, B. Inflammatory cytokines and acute graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation. Blood 2005, 106, 4407–4411. [Google Scholar] [CrossRef] [Green Version]
  26. Nimer, S.D.; Giorgi, J.; Gajewski, J.L.; Ku, N.; Schiller, G.J.; Lee, K.; Territo, M.; Ho, W.; Feio, S.; Selch, M.; et al. Selective depletion of CD8+ cells for prevention of graft-versus-host disease after bone marrow transplantation. A randomized controlled trial. Transplantation 1994, 57, 82–87. [Google Scholar] [CrossRef]
  27. Via, C.S.; Finkelman, F.D. Critical role of interleukin-2 in the development of acute graft-versus-host disease. Int. Immunol. 1993, 5, 565–572. [Google Scholar] [CrossRef]
  28. Shustov, A.; Nguyen, P.; Finkelman, F.; Elkon, K.B.; Via, C.S. Related Content Fas Expression on Antigen-Speciic T Cells Has Costimulatory, Helper, and Down-Regulatory Functions In Vivo for Cytotoxic T Cell Responses but Not for T Cell-Dependent B Cell Responses. J. Immunol. 1998, 161, 2848–2855. [Google Scholar] [CrossRef]
  29. Koyama, M.; Cheong, M.; Markey, K.A.; Gartlan, K.; Kuns, R.D.; Locke, K.R.; Lineburg, K.E.; Teal, B.E.; Mouttie, L.L.-E.; Bunting, M.D.; et al. Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease. J. Exp. Med. 2015, 212, 1303–1321. [Google Scholar] [CrossRef] [Green Version]
  30. Ghayur, T.; Seemayer, T.A.; Kongshavn, P.A.; Gartner, J.G.; Lapp, W.S. Graft-versus-host reactions in the beige mouse. An investigation of the role of host and donor natural killer cells in the pathogenesis of graft-versus-host disease. Transplantation 1987, 44, 261–267. [Google Scholar] [CrossRef]
  31. Ruggeri, L.; Aversa, F.; Martelli, M.F.; Velardi, A. Allogeneic hematopoietic transplantation and natural killer cell recognition of missing self. Immunol. Rev. 2006, 214, 202–218. [Google Scholar] [CrossRef] [PubMed]
  32. Ball, L.M.; Egeler, R.M. Acute GvHD: Pathogenesis and classification. Bone Marrow Transplant. 2008, 41, S58–S64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Arai, S.; Arora, M.; Wang, T.; Spellman, S.R.; He, W.; Couriel, D.R.; Urbano-Ispizua, A.; Cutler, C.S.; Bacigalupo, A.A.; Battiwalla, M.; et al. Increasing Incidence of Chronic Graft-versus-Host Disease inAllogeneic Transplantation: A Report from the Center for International Blood and Marrow Transplant Research. Biol. Blood Marrow Transplant. 2015, 21, 266–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Flowers, M.E.D.; Parker, P.M.; Johnston, L.J.; Matos, A.V.B.; Storer, B.; Bensinger, W.I.; Storb, R.; Appelbaum, F.R.; Forman, S.J.; Blume, K.G.; et al. Comparison of chronic graft-versus-host disease after transplantation of peripheral blood stem cells versus bone marrow in allogeneic recipients: Long-term follow-up of a randomized trial. Blood 2002, 100, 415–419. [Google Scholar] [CrossRef] [PubMed]
  35. Cooke, K.R.; Luznik, L.; Sarantopoulos, S.; Hakim, F.T.; Jagasia, M.; Fowler, D.H.; Brink, M.R.V.D.; Hansen, J.A.; Parkman, R.; Miklos, D.B.; et al. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2017, 23, 211–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Martires, K.J.; Baird, K.; Citrin, D.E.; Hakim, F.T.; Pavletic, S.Z.; Cowen, E.W. Localization of sclerotic-type chronic graft-vs-host disease to sites of skin injury: Potential insight into the mechanism of isomorphic and isotopic responses. Arch. Dermatol. 2011, 147, 1081–1086. [Google Scholar] [CrossRef] [Green Version]
  37. Nestel, F.P.; Price, K.S.; Seemayer, T.A.; Lapp, W.S. Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor alpha during graft-versus-host disease. J. Exp. Med. 1992, 175, 405–413. [Google Scholar] [CrossRef] [Green Version]
  38. Rangarajan, H.; Yassai, M.; Subramanian, H.; Komorowski, R.; Whitaker, M.; Gorski, J.; Drobyski, W.R. Emergence of T Cells that recognize nonpolymorphic antigens during graft-versus-host disease. Blood 2012, 119, 6354–6364. [Google Scholar] [CrossRef]
  39. Van Bekkum, D.W.; Knaan, S. Role of bacterial microflora in development of intestinal lesions from graft-versus-host reaction. J. Natl. Cancer Inst. 1977, 58, 787–790. [Google Scholar] [CrossRef]
  40. Brüggen, M.C.; Klein, I.; Greinix, H.; Bauer, W.; Kuzmina, Z.; Rabitsch, W.; Kalhs, P.; Petzelbauer, P.; Knobler, R.; Stingl, G.; et al. Diverse T-cell responses characterize the different manifestations of cutaneous graft-versus-host disease. Blood 2014, 123, 290–299. [Google Scholar] [CrossRef] [Green Version]
  41. Fujiwara, H.; Maeda, Y.; Kobayashi, K.; Nishimori, H.; Matsuoka, K.-I.; Fujii, N.; Kondo, E.; Tanaka, T.; Chen, L.; Azuma, M.; et al. Programmed death-1 pathway in host tissues ameliorates Th17/Th1-mediated experimental chronic graft-versus-host disease. J. Immunol. 2014, 193, 2565–2573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Malard, F.; Bossard, C.; Brissot, E.; Chevallier, P.; Guillaume, T.; Delaunay, J.; Mosnier, J.-F.; Moreau, P.; Grégoire, M.; Gaugler, B.; et al. Increased Th17/Treg ratio in chronic liver GVHD. Bone Marrow Transpl. 2014, 49, 539–544. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, Y.; Huang, L.; Huang, T.; Geng, S.; Chen, X.; Huang, X.; Lai, P.; Du, X.; Weng, J. The Gut Bacteria Dysbiosis Contributes to Chronic Graft-versus-Host Disease Associated with a Treg/Th1 Ratio Imbalance. Front. Microbiol. 2022, 13, 813576. [Google Scholar] [CrossRef] [PubMed]
  44. Dudakov, J.A.; Mertelsmann, A.M.; O’connor, M.H.; Jenq, R.R.; Velardi, E.; Young, L.F.; Smith, O.M.; Boyd, R.L.; Brink, M.R.M.V.D.; Hanash, A.M. Loss of thymic innate lymphoid cells leads to impaired thymopoiesis in experimental graft-versus-host disease. Blood 2017, 130, 933–942. [Google Scholar] [CrossRef] [Green Version]
  45. Fukushi, N.; Arase, H.; Wang, B.; Ogasawara, K.; Gotohda, T.; Good, R.A.; Onoé, K. Thymus: A direct target tissue in graft-versus-host reaction after allogeneic bone marrow transplantation that results in abrogation of induction of self-tolerance. Proc. Natl. Acad. Sci. USA 1990, 87, 6301–6305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Hauri-Hohl, M.M.; Keller, M.P.; Gill, J.; Hafen, K.; Pachlatko, E.; Boulay, T.; Peter, A.; Holländer, G.A.; Krenger, W. Donor T-cell alloreactivity against host thymic epithelium limits T-cell development after bone marrow transplantation. Blood 2007, 109, 4080–4088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Sakoda, Y.; Hashimoto, D.; Asakura, S.; Takeuchi, K.; Harada, M.; Tanimoto, M.; Teshima, T. Donor-derived thymic-dependent T Cells cause chronic graft-versus-host disease. Blood 2007, 109, 1756–1764. [Google Scholar] [CrossRef] [Green Version]
  48. Teshima, T.; Reddy, P.; Liu, C.; Williams, D.; Cooke, K.R.; Ferrara, J.L.M. Impaired thymic negative selection causes autoimmune graft-versus-host disease. Blood 2003, 102, 429–435. [Google Scholar] [CrossRef] [Green Version]
  49. Wu, T.; Young, J.S.; Johnston, H.; Ni, X.; Deng, R.; Racine, J.; Wang, M.; Wang, A.; Todorov, I.; Wang, J.; et al. Thymic damage, impaired negative selection, and development of chronic graft-versus-host disease caused by donor CD4+ and CD8+ T Cells. J. Immunol. 2013, 191, 488–499. [Google Scholar] [CrossRef] [Green Version]
  50. Zhao, D.; Young, J.S.; Chen, Y.-H.; Shen, E.; Yi, T.; Todorov, I.; Chu, P.G.; Forman, S.J.; Zeng, D. Alloimmune response results in expansion of autoreactive donor CD4+ T Cells in transplants that can mediate chronic graft-versus-host disease. J. Immunol. 2011, 186, 856–868. [Google Scholar] [CrossRef] [Green Version]
  51. Alho, A.C.; Kim, H.T.; Chammas, M.J.; Reynolds, C.G.; Matos, T.R.; Forcade, E.; Whangbo, J.; Nikiforow, S.; Cutler, C.S.; Koreth, J.; et al. Unbalanced recovery of regulatory and effector T Cells after allogeneic stem cell transplantation contributes to chronic GVHD. Blood 2016, 127, 646–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Allen, J.L.; Fore, M.S.; Wooten, J.; Roehrs, P.A.; Bhuiya, N.S.; Hoffert, T.; Sharf, A.; Deal, A.M.; Armistead, P.; Coghill, J.; et al. B cells from patients with chronic GVHD are activated and primed for survival via BAFF-mediated pathways. Blood 2012, 120, 2529–2536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Sarantopoulos, S.; Stevenson, K.E.; Kim, H.T.; Bhuiya, N.S.; Cutler, C.S.; Soiffer, R.J.; Antin, J.H.; Ritz, J. High levels of B-cell activating factor in patients with active chronic graft-versus-host disease. Clin. Cancer Res. 2007, 13, 6107–6114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Sarantopoulos, S.; Stevenson, K.E.; Kim, H.T.; Cutler, C.S.; Bhuiya, N.S.; Schowalter, M.; Ho, V.T.; Alyea, E.P.; Koreth, J.; Blazar, B.R.; et al. Altered B-cell homeostasis and excess BAFF in human chronic graft-versus-host disease. Blood 2009, 113, 3865–3874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Shen, Z.; Ma, J.; Peng, R.; Hu, B.; Zhao, Y.; Liu, S.; Hong, J. Biomarkers in Ocular Graft-Versus-Host Disease: Implications for the Involvement of B Cells. Transpl. Cell Ther. 2022, 28, e1–e749. [Google Scholar] [CrossRef]
  56. Wan, L.; Jin, Z.; Hu, B.; Lv, K.; Lei, L.; Liu, Y.; Song, Y.; Zhu, Y.; Gong, H.; Xu, M.; et al. IL-Y Aggravates Murine Chronic Graft- Versus-Host Disease by Enhancing T and B Cell Responses. Front. Immunol. 2020, 11, 559740. [Google Scholar] [CrossRef]
  57. Zhai, Z.; Sun, Z.; Li, Q.; Zhang, A.; Liu, H.; Xu, J.; Xu, X.; Geng, L.; Harris, D.; Hu, S.; et al. Correlation of the CD4+CD25high T-regulatory cells in recipients and their corresponding donors to acute GVHD. Transpl. Int. 2007, 20, 440–446. [Google Scholar] [CrossRef]
  58. Alexander, K.A.; Flynn, R.; Lineburg, K.E.; Kuns, R.D.; Teal, B.E.; Olver, S.D.; Lor, M.; Raffelt, N.C.; Koyama, M.; Leveque, L.; et al. CSF-1-dependant donor-derived macrophages mediate chronic graft-versus-host disease. J. Clin. Investig. 2014, 124, 4266–4280. [Google Scholar] [CrossRef] [Green Version]
  59. Du, J.; Paz, K.; Flynn, R.; Vulic, A.; Robinson, T.M.; Lineburg, K.E.; Alexander, K.A.; Meng, J.; Roy, S.; Panoskaltsis-Mortari, A.; et al. Pirfenidone ameliorates murine chronic GVHD through inhibition of macrophage infiltration and TGF-β production. Blood 2017, 129, 2570–2580. [Google Scholar] [CrossRef] [Green Version]
  60. Flynn, R.; Du, J.; Veenstra, R.G.; Reichenbach, D.K.; Panoskaltsis-Mortari, A.; Taylor, P.A.; Freeman, G.J.; Serody, J.; Murphy, W.J.; Munn, D.; et al. Increased T follicular helper cells and germinal center B cells are required for cGVHD and bronchiolitis obliterans. Blood 2014, 123, 3988–3998. [Google Scholar] [CrossRef] [Green Version]
  61. Guo, L.; Lai, P.; Wang, Y.; Huang, T.; Chen, X.; Geng, S.; Huang, X.; Luo, C.; Wu, S.; Ling, W.; et al. Extracellular vesicles derived from mesenchymal stem cells prevent skin fibrosis in the cGVHD mouse model by suppressing the activation of macrophages and B cells immune response. Int. Immunopharmacol. 2020, 84, 106541. [Google Scholar] [CrossRef] [PubMed]
  62. Kuroi, T.; Fujii, N.; Ichimura, K.; Seike, K.; Yamamoto, A.; Kambara, Y.; Sugimoto, S.; Otani, S.; Saeki, K.; Fujiwara, H.; et al. Characterization of localized macrophages in bronchiolitis obliterans after allogeneic hematopoietic cell transplantation. Int. J. Hematol. 2021, 114, 701–708. [Google Scholar] [CrossRef] [PubMed]
  63. Panoskaltsis-Mortari, A.; Tram, K.V.; Price, A.P.; Wendt, C.H.; Blazar, B.R. A new murine model for bronchiolitis obliterans post-bone marrow transplant. Am. J. Respir. Crit. Care Med. 2007, 176, 713–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Srinivasan, M.; Flynn, R.; Price, A.; Ranger, A.; Browning, J.L.; Taylor, P.A.; Ritz, J.; Antin, J.H.; Murphy, W.J.; Luznik, L.; et al. Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans. Blood 2012, 119, 1570–1580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chaturvedi, S.; George, B.; Savani, B.N. Bleeding and thrombotic complications. In The EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies; Springer: Berlin/Heidelberg, Germany, 2018; pp. 301–306. [Google Scholar] [CrossRef]
  66. Shatry, A.M.; Roopenian, D.C.; Levy, R.B. Survival and function of MiHA epitope-specific host CD8 TM cells following ablative conditioning and HCT. Biol. Blood Marrow Transpl. 2007, 13, 293–298. [Google Scholar] [CrossRef] [Green Version]
  67. Marijt, W.A.; Kernan, N.A.; Diaz-Barrientos, T.; Veenhof, W.F.; O’Reilly, R.J.; Willemze, R.; Falkenburg, J.H. Multiple minor histocompatibility antigen-specific cytotoxic T lymphocyte clones can be generated during graft rejection after HLA-identical bone marrow transplantation. Bone Marrow Transpl. 1995, 16, 125–132. [Google Scholar]
  68. Marijt, W.A.; Veenhof, W.F.; Brand, A.; Goulmy, E.; Fibbe, W.E.; Willemze, R.; van Rood, J.J.; Falkenburg, J.H. Minor histocompatibility antigen-specific cytotoxic T Cell lines, capable of lysing human hematopoietic progenitor cells, can be generated in vitro by stimulation with HLA-identical bone marrow cells. J. Exp. Med. 1991, 173, 101–109. [Google Scholar] [CrossRef] [Green Version]
  69. Taylor, M.A.; Ward, B.; Schatzle, J.D.; Bennett, M. Perforin- and Fas-dependent mechanisms of natural killer cell-mediated rejection of incompatible bone marrow cell grafts. Eur. J. Immunol. 2002, 32, 793–799. [Google Scholar] [CrossRef]
  70. Bennett, M.; Taylor, P.A.; Austin, M.; Baker, M.B.; Schook, L.; Rutherford, M.S.; Kumar, V.; Podack, E.R.; Mohler, K.M.; Levy, R.B.; et al. Cytokine and cytotoxic pathways of NK cell rejection of class I-deficient bone marrow grafts: Influence of mouse colony environment. Int. Immunol. 1998, 10, 785–790. [Google Scholar] [CrossRef] [Green Version]
  71. Zimmerman, Z.; Shatry, A.; Deyev, V.; Podack, E.; Mammolenti, M.; Blazar, B.R.; Yagita, H.; Levy, R.B. Effector cells derived from host CD8 memory T Cells mediate rapid resistance against minor histocompatibility antigen-mismatched allogeneic marrow grafts without participation of perforin, Fas ligand, and the simultaneous inhibition of 3 tumor necrosis factor family effector pathways. Biol. Blood Marrow Transpl. 2005, 11, 576–586. [Google Scholar] [CrossRef] [Green Version]
  72. Komatsu, M.; Mammolenti, M.; Jones, M.; Jurecic, R.; Sayers, T.J.; Levy, R.B. Antigen-primed CD8+ T Cells can mediate resistance, preventing allogeneic marrow engraftment in the simultaneous absence of perforin-, CD95L-, TNFR1-, and TRAIL-dependent killing. Blood 2003, 101, 3991–3999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Merli, P.; Caruana, I.; De Vito, R.; Strocchio, L.; Weber, G.; Del Bufalo, F.; Buatois, V.; Montanari, P.; Cefalo, M.G.; Pitisci, A.; et al. Role of interferon-γ in immune-mediated graft failure after allogeneic hematopoietic stem cell transplantation. Haematologica 2019, 104, 2314–2323. [Google Scholar] [CrossRef] [PubMed]
  74. Koyama, M.; Hashimoto, D.; Nagafuji, K.; Eto, T.; Ohno, Y.; Aoyama, K.; Iwasaki, H.; Miyamoto, T.; Hill, G.; Akashi, K.; et al. Expansion of donor-reactive host T Cells in primary graft failure after allogeneic hematopoietic SCT following reduced-intensity conditioning. Bone Marrow Transpl. 2014, 49, 110–115. [Google Scholar] [CrossRef] [Green Version]
  75. Lapidot, T.; Faktorowich, Y.; Lubin, I.; Reisner, Y. Enhancement of T-cell-depleted bone marrow allografts in the absence of graft-versus-host disease is mediated by CD8+ CD4− and not by CD8- CD4+ thymocytes. Blood 1992, 80, 2406–2411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Martin, P.J. Prevention of allogeneic marrow graft rejection by donor T Cells that do not recognize recipient alloantigens: Potential role of a veto mechanism. Blood 1996, 88, 962–969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Martin, P.J. Donor CD8 cells prevent allogeneic marrow graft rejection in mice: Potential implications for marrow transplantation in humans. J. Exp. Med. 1993, 178, 703–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Murphy, W.J.; Bennett, M.; Kumar, V.; Longo, D.L. Donor-type activated natural killer cells promote marrow engraftment and B cell development during allogeneic bone marrow transplantation. J. Immunol. 1992, 148, 2953–2960. [Google Scholar] [CrossRef]
  79. Fujisaki, J.; Wu, J.; Carlson, A.L.; Silberstein, L.; Putheti, P.; Larocca, R.; Gao, W.; Saito, T.I.; Celso, C.L.; Tsuyuzaki, H.; et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature 2011, 474, 216–219. [Google Scholar] [CrossRef] [Green Version]
  80. Joffre, O.; Santolaria, T.; Calise, D.; Al Saati, T.; Hudrisier, D.; Romagnoli, P.; Van Meerwijk, J.P.M. Prevention of acute and chronic allograft rejection with CD4+CD25+Foxp3+ regulatory T lymphocytes. Nat. Med. 2008, 14, 88–92. [Google Scholar] [CrossRef] [Green Version]
  81. Barao, I.; Hanash, A.M.; Hallett, W.; Welniak, L.A.; Sun, K.; Redelman, D.; Blazar, B.R.; Levy, R.B.; Murphy, W.J. Suppression of natural killer cell-mediated bone marrow cell rejection by CD4+CD25+ regulatory T Cells. Proc. Natl. Acad. Sci. USA 2006, 103, 5460–5465. [Google Scholar] [CrossRef] [Green Version]
  82. Park, J.H.; Lee, J.-H.; Lee, J.-H.; Park, H.-S.; Choi, E.-J.; Kang, Y.-A.; Kang, H.; Woo, J.M.; Lee, Y.-S.; Jeon, M.; et al. Incidence, Management, and Prognosis of Graft Failure and Autologous Reconstitution after Allogeneic Hematopoietic Stem Cell Transplantation. J. Korean Med. Sci. 2021, 36, e151. [Google Scholar] [CrossRef] [PubMed]
  83. Servais, S.; Beguin, Y.; Baron, F. Emerging drugs for prevention of graft failure after allogeneic hematopoietic stem cell transplantation. Expert Opin. Emerg. Drugs 2013, 18, 173–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Astor, B.C.; Melamed, M.L.; Mandelbrot, D.A.; Djamali, A. Seasonality of mortality and graft failure among kidney transplant recipients in the US—A retrospective study. Transpl. Int. 2018, 31, 293–301. [Google Scholar] [CrossRef] [Green Version]
  85. Tinckam, K. Histocompatibility methods. Transpl. Rev. 2009, 23, 80–93. [Google Scholar] [CrossRef] [PubMed]
  86. Cai, J.; Terasaki, P.I. Humoral theory of transplantation: Mechanism, prevention, and treatment. Hum. Immunol. 2005, 66, 334–342. [Google Scholar] [CrossRef] [PubMed]
  87. Lentine, K.L.; Smith, J.; Hart, A.; Miller, J.; Skeans, M.; Larkin, L.; Robinson, A.; Gauntt, K.; Israni, A.; Hirose, R.; et al. OPTN/SRTR 2020 Annual Data Report: Kidney. Am. J. Transpl. 2022, 22 (Suppl. 2), 21–136. [Google Scholar] [CrossRef] [PubMed]
  88. Kloc, M.; Ghobrial, R.M. Chronic allograft rejection: A significant hurdle to transplant success. Burn. Trauma 2014, 2, 3–10. [Google Scholar] [CrossRef] [Green Version]
  89. Bestard, O.; Nickel, P.; Cruzado, J.M.; Schoenemann, C.; Boenisch, O.; Sefrin, A.; Grinyó, J.M.; Volk, H.-D.; Reinke, P. Circulating Alloreactive T Cells Correlate with Graft Function in Longstanding Renal Transplant Recipients. J. Am. Soc. Nephrol. 2008, 19, 1419–1429. [Google Scholar] [CrossRef] [Green Version]
  90. Kishimoto, K.; Sandner, S.; Imitola, J.; Sho, M.; Li, Y.; Langmuir, P.B.; Rothstein, D.M.; Strom, T.B.; Turka, L.A.; Sayegh, M.H. Th1 cytokines, programmed cell death, and alloreactive T Cell clone size in transplant tolerance. J. Clin. Investig. 2002, 109, 1471–1479. [Google Scholar] [CrossRef]
  91. Kythreotou, A.; Siddique, A.; Mauri, F.A.; Bower, M.; Pinato, D.J. PD-L1. J. Clin. Pathol. 2018, 71, 189–194. [Google Scholar] [CrossRef]
  92. Liang, S.C.; Latchman, Y.E.; Buhlmann, J.E.; Tomczak, M.F.; Horwitz, B.H.; Freeman, G.J.; Sharpe, A.H. Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses. Eur. J. Immunol. 2003, 33, 2706–2716. [Google Scholar] [CrossRef] [PubMed]
  93. Dong, H.; Zhu, G.; Tamada, K.; Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 1999, 5, 1365–1369. [Google Scholar] [CrossRef] [PubMed]
  94. Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992, 11, 3887–3895. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, L.; Flies, D.B. Molecular mechanisms of T Cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef]
  96. Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 2010, 236, 219–242. [Google Scholar] [CrossRef]
  97. Pesce, S.; Greppi, M.; Tabellini, G.; Rampinelli, F.; Parolini, S.; Olive, D.; Moretta, L.; Moretta, A.; Marcenaro, E. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J. Allergy Clin. Immunol. 2017, 139, 335–346.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Zhulai, G.; Oleinik, E. Targeting regulatory T cells in anti-PD-1/PD-L1 cancer immunotherapy. Scan. J. Immunol. 2022, 95, e13129. [Google Scholar]
  99. Francisco, L.M.; Salinas, V.H.; Brown, K.E.; Vanguri, V.K.; Freeman, G.J.; Kuchroo, V.K.; Sharpe, A.H. PD-L1 regulates the development, maintenance, and function of induced regulatory T Cells. J. Exp. Med. 2009, 206, 3015–3029. [Google Scholar] [CrossRef] [Green Version]
  100. Bretscher, P.A. A two-step, two-signal model for the primary activation of precursor helper T Cells. Proc. Natl. Acad. Sci. USA 1999, 96, 185–190. [Google Scholar] [CrossRef] [Green Version]
  101. Patsoukis, N.; Duke-Cohan, J.S.; Chaudhri, A.; Aksoylar, H.-I.; Wang, Q.; Council, A.; Berg, A.; Freeman, G.J.; Boussiotis, V.A. Interaction of SHP-2 SH2 domains with PD-1 ITSM induces PD-1 dimerization and SHP-2 activation. Commun. Biol. 2020, 3, 128. [Google Scholar] [CrossRef] [Green Version]
  102. Chikuma, S.; Terawaki, S.; Hayashi, T.; Nabeshima, R.; Yoshida, T.; Shibayama, S.; Okazaki, T.; Honjo, T. PD-1-mediated suppression of IL-2 production induces CD8+ T Cell anergy in vivo. J. Immunol. 2009, 182, 6682–6689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Boussiotis, V.A.; Chatterjee, P.; Li, L. Biochemical signaling of PD-1 on T Cells and its functional implications. Cancer J. 2014, 20, 265–271. [Google Scholar] [CrossRef]
  104. Yokosuka, T.; Takamatsu, M.; Kobayashi-Imanishi, W.; Hashimoto-Tane, A.; Azuma, M.; Saito, T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T Cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 2012, 209, 1201–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Appleman, L.J.; van Puijenbroek, A.A.F.L.; Shu, K.M.; Nadler, L.M.; Boussiotis, V.A. CD28 costimulation mediates down-regulation of p27kip1 and cell cycle progression by activation of the PI3K/PKB signaling pathway in primary human T Cells. J. Immunol. 2002, 168, 2729–2736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Riley, J.L. PD-1 signaling in primary T Cells. Immunol. Rev. 2009, 229, 114–125. [Google Scholar] [CrossRef] [Green Version]
  107. Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef] [Green Version]
  108. Pompura, S.L.; Dominguez-Villar, M. The PI3K/AKT signaling pathway in regulatory T-cell development, stability, and function. J. Leukoc. Biol 2018, 103, 1065–1076. [Google Scholar] [CrossRef]
  109. Patsoukis, N.; Sari, D.; Boussiotis, V.A. PD-1 inhibits T Cell proliferation by upregulating p27 and p15 and suppressing Cdc25A. Cell Cycle 2012, 11, 4305–4309. [Google Scholar] [CrossRef] [Green Version]
  110. Zhao, Y.; Lee, C.K.; Lin, C.-H.; Gassen, R.B.; Xu, X.; Huang, Z.; Xiao, C.; Bonorino, C.; Lu, L.-F.; Bui, J.D.; et al. PD-L1:CD80 Cis-Heterodimer Triggers the Co-stimulatory Receptor CD28 While Repressing the Inhibitory PD-1 and CTLA-4 Pathways. Immunity 2019, 51, 1059–1073.e9. [Google Scholar] [CrossRef]
  111. Deng, R.; Cassady, K.; Li, X.; Yao, S.; Zhang, M.; Racine, J.; Lin, J.; Chen, L.; Zeng, D. B7H1/CD80 interaction augments PD-1-dependent T Cell apoptosis and ameliorates graft-versus-host disease. J. Immunol. 2015, 194, 560–574. [Google Scholar] [CrossRef] [Green Version]
  112. Sugiura, D.; Maruhashi, T.; Okazaki, I.-M.; Shimizu, K.; Maeda, T.K.; Takemoto, T.; Okazaki, T. Restriction of PD-1 function by cis-PD-L1/CD80 interactions is required for optimal T Cell responses. Science 2019, 364, 558–566. [Google Scholar] [CrossRef] [PubMed]
  113. Yi, T.; Li, X.; Yao, S.; Wang, L.; Chen, Y.; Zhao, D.; Johnston, H.F.; Young, J.S.; Liu, H.; Todorov, I.; et al. Host APCs augment in vivo expansion of donor natural regulatory T Cells via B7H1/B7.1 in allogeneic recipients. J. Immunol. 2011, 186, 2739–2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Park, J.-J.; Omiya, R.; Matsumura, Y.; Sakoda, Y.; Kuramasu, A.; Augustine, M.M.; Yao, S.; Tsushima, F.; Narazaki, H.; Anand, S.; et al. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood 2010, 116, 1291–1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Rollins, M.R.; Johnson, R.M.G. CD80 Expressed by CD8+ T Cells Contributes to PD-L1-Induced Apoptosis of Activated CD8+ T Cells. J. Immunol. Res. 2017, 2017, 7659462. [Google Scholar] [CrossRef] [Green Version]
  116. Tang, L.; Ma, S.; Gong, H.; Wang, J.; Xu, Y.; Wu, D.; Sun, A. PD-L1 Ameliorates Murine Acute Graft-versus-Host Disease by Suppressing Effector but Not Regulatory T Cells Function. Arch. Immunol. Ther. Exp. 2019, 67, 179–187. [Google Scholar] [CrossRef]
  117. Blazar, B.R.; Carreno, B.M.; Panoskaltsis-Mortari, A.; Carter, L.; Iwai, Y.; Yagita, H.; Nishimura, H.; Taylor, P.A. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J. Immunol. 2003, 171, 1272–1277. [Google Scholar] [CrossRef] [Green Version]
  118. Li, X.; Deng, R.; He, W.; Liu, C.; Wang, M.; Young, J.; Meng, Z.; Du, C.; Huang, W.; Chen, L.; et al. Loss of B7-H1 expression by recipient parenchymal cells leads to expansion of infiltrating donor CD8+ T Cells and persistence of graft-versus-host disease. J. Immunol. 2012, 188, 724–734. [Google Scholar] [CrossRef] [Green Version]
  119. Saha, A.; O’connor, R.S.; Thangavelu, G.; Lovitch, S.; Dandamudi, D.B.; Wilson, C.B.; Vincent, B.G.; Tkachev, V.; Pawlicki, J.M.; Furlan, S.; et al. Programmed death ligand-1 expression on donor T Cells drives graft-versus-host disease lethality. J. Clin. Investig. 2016, 126, 2642–2660. [Google Scholar] [CrossRef]
  120. Kinter, A.L.; Godbout, E.J.; McNally, J.P.; Sereti, I.; Roby, G.A.; O’Shea, M.A.; Fauci, A.S. The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J. Immunol. 2008, 181, 6738–6746. [Google Scholar] [CrossRef] [Green Version]
  121. Hu, Q.-Y.; Yuan, Y.; Li, Y.-C.; Yang, L.-Y.; Zhou, X.-Y.; Xiong, D.-Q.; Zhao, Z.-Y. Programmed Cell Death Ligand 1-Transfected Mouse Bone Marrow Mesenchymal Stem Cells as Targeted Therapy for Rheumatoid Arthritis. BioMed Res. Int. 2021, 2021, 5574282. [Google Scholar] [CrossRef]
  122. Sugiura, D.; Okazaki, I.-M.; Maeda, T.K.; Maruhashi, T.; Shimizu, K.; Arakaki, R.; Takemoto, T.; Ishimaru, N.; Okazaki, T. PD-1 agonism by anti-CD80 inhibits T Cell activation and alleviates autoimmunity. Nat. Immunol. 2022, 23, 399–410. [Google Scholar] [CrossRef]
  123. Nishimura, H.; Okazaki, T.; Tanaka, Y.; Nakatani, K.; Hara, M.; Matsumori, A.; Sasayama, S.; Mizoguchi, A.; Hiai, H.; Minato, N.; et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001, 291, 319–322. [Google Scholar] [CrossRef] [PubMed]
  124. Latchman, Y.E.; Liang, S.C.; Wu, Y.; Chernova, T.; Sobel, R.A.; Klemm, M.; Kuchroo, V.K.; Freeman, G.J.; Sharpe, A.H. PD-L1-deficient mice show that PD-L1 on T Cells, antigen-presenting cells, and host tissues negatively regulates T Cells. Proc. Natl. Acad. Sci. USA 2004, 101, 10691–10696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Kasagi, S.; Kawano, S.; Okazaki, T.; Honjo, T.; Morinobu, A.; Hatachi, S.; Shimatani, K.; Tanaka, Y.; Minato, N.; Kumagai, S. Anti-programmed cell death 1 antibody reduces CD4+PD-1+ T Cells and relieves the lupus-like nephritis of NZB/W F1 mice. J. Immunol. 2010, 184, 2337–2347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Tanaka, K.; Albin, M.J.; Yuan, X.; Yamaura, K.; Habicht, A.; Murayama, T.; Grimm, M.; Waaga, A.M.; Ueno, T.; Padera, R.F.; et al. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. J. Immunol. 2007, 179, 5204–5210. [Google Scholar] [CrossRef] [Green Version]
  127. Morita, M.; Fujino, M.; Jiang, G.; Kitazawa, Y.; Xie, L.; Azuma, M.; Yagita, H.; Nagao, S.; Sugioka, A.; Kurosawa, Y.; et al. PD-1/B7-H1 interaction contribute to the spontaneous acceptance of mouse liver allograft. Am. J. Transpl. 2010, 10, 40–46. [Google Scholar] [CrossRef] [Green Version]
  128. Tokita, D.; Mazariegos, G.V.; Zahorchak, A.F.; Chien, N.; Abe, M.; Raimondi, G.; Thomson, A.W. High PD-L1/CD86 ratio on plasmacytoid dendritic cells correlates with elevated T-regulatory cells in liver transplant tolerance. Transplantation 2008, 85, 369–377. [Google Scholar] [CrossRef]
  129. Cha, J.-H.; Chan, L.-C.; Li, C.-W.; Hsu, J.L.; Hung, M.-C. Mechanisms Controlling PD-L1 Expression in Cancer. Mol. Cell 2019, 76, 359–370. [Google Scholar] [CrossRef]
  130. Ai, L.; Xu, A.; Xu, J. Roles of PD-1/PD-L1 Pathway: Signaling, Cancer, and Beyond. Adv. Exp. Med. Biol. 2020, 1248, 33–59. [Google Scholar] [CrossRef]
  131. Hall, K.H.; Liu, Y.; Jiang, C.; Harvey, R.D. New and Worsening Long-term Immune-Related Adverse Events with PD-1/PD-L1 Pathway Agents in Patients with Cancer. Pharmacotherapy 2020, 40, 133–141. [Google Scholar] [CrossRef]
  132. Delanoy, N.; Michot, J.-M.; Comont, T.; Kramkimel, N.; Lazarovici, J.; Dupont, R.; Champiat, S.; Chahine, C.; Robert, C.; Herbaux, C.; et al. Haematological immune-related adverse events induced by anti-PD-1 or anti-PD-L1 immunotherapy: A descriptive observational study. Lancet Haematol 2019, 6, e48–e57. [Google Scholar] [CrossRef] [PubMed]
  133. Canavan, M.; Floudas, A.; Veale, D.J.; Fearon, U. The PD-1:PD-L1 axis in Inflammatory Arthritis. BMC Rheumatol. 2021, 5, 1. [Google Scholar] [CrossRef] [PubMed]
  134. Wu, Q.; Jiang, L.; Li, S.-C.; He, Q.-J.; Yang, B.; Cao, J. Small molecule inhibitors targeting the PD-1/PD-L1 signaling pathway. Acta Pharmacol. Sin. 2021, 42, 1–9. [Google Scholar] [CrossRef] [PubMed]
  135. Xiong, W.; Gao, Y.; Wei, W.; Zhang, J. Extracellular and nuclear PD-L1 in modulating cancer immunotherapy. Trends Cancer 2021, 7, 837–846. [Google Scholar] [CrossRef]
  136. Ying, H.; Zhang, X.; Duan, Y.; Lao, M.; Xu, J.; Yang, H.; Liang, T.; Bai, X. Non-cytomembrane PD-L1: An atypical target for cancer. Pharmacol. Res. 2021, 170, 105741. [Google Scholar] [CrossRef]
  137. Yi, M.; Niu, M.; Xu, L.; Luo, S.; Wu, K. Regulation of PD-L1 expression in the tumor microenvironment. J. Hematol. Oncol. 2021, 14, 10. [Google Scholar] [CrossRef]
  138. Elalouf, A. Infections after organ transplantation and immune response. Transpl. Immunol. 2023, 77, 101798. [Google Scholar] [CrossRef]
  139. Vietzen, H.; Furlano, P.L.; Cornelissen, J.J.; Böhmig, G.A.; Jaksch, P.; Puchhammer-Stöckl, E. HLA-E-restricted immune responses are crucial for the control of EBV infections and the prevention of PTLD. Blood 2023, 141, 1560–1573. [Google Scholar] [CrossRef]
  140. Park, J.-I.; Song, G.-W.; Ryu, J.H.; Choi, S.-T.; Choi, N.-G.; Jung, B.-H.; Chu, C.W.; Kim, K.-K.; Jung, D.-H.; Ha, T.-Y.; et al. A Multicenter, Randomized, Open-Label Study to Compare the Efficacy and Safety of Tacrolimus and Corticosteroids in Combination with or without Mycophenolate Mofetil in Liver Transplantation Recipients Infected with Hepatitis B Virus. Transplant Proc. 2023, 55, 387–395. [Google Scholar] [CrossRef]
  141. Burek Kamenaric, M.; Ivkovic, V.; Kovacevic Vojtusek, I.; Zunec, R. The Role of HLA and KIR Immunogenetics in BK Virus Infection after Kidney Transplantation. Viruses 2020, 12, 1417. [Google Scholar] [CrossRef]
Figure 1. A DC acts as an APC for both CD8+ and CD4+ T cells via MHC I and MHCII, respectively, allowing for co-stimulatory/inhibitory signaling to occur. (a) MHC presentation of a complimentary peptide to the TCR allows for CD80’s activation of CD28 on the T cell, which subsequently enhances T cell metabolism and proliferation via the PI3K/AKT pathway. PD-L1 is sequestered by CD80, forming a heterodimer. CD80 simultaneously blocks the activation of PD-1 while activating CD28. (b) MHC presentation of a complimentary peptide to the TCR, followed by PD-L1 recognition by PD-1, induces phosphorylation of ITSM on the intracellular portion of PD-1. Phosphorylated ITSM then interacts with SHP-2, inhibiting the PIK3/AKT signaling cascade and limiting T cell production of IL-2.
Figure 1. A DC acts as an APC for both CD8+ and CD4+ T cells via MHC I and MHCII, respectively, allowing for co-stimulatory/inhibitory signaling to occur. (a) MHC presentation of a complimentary peptide to the TCR allows for CD80’s activation of CD28 on the T cell, which subsequently enhances T cell metabolism and proliferation via the PI3K/AKT pathway. PD-L1 is sequestered by CD80, forming a heterodimer. CD80 simultaneously blocks the activation of PD-1 while activating CD28. (b) MHC presentation of a complimentary peptide to the TCR, followed by PD-L1 recognition by PD-1, induces phosphorylation of ITSM on the intracellular portion of PD-1. Phosphorylated ITSM then interacts with SHP-2, inhibiting the PIK3/AKT signaling cascade and limiting T cell production of IL-2.
Cells 12 01609 g001aCells 12 01609 g001b
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Handelsman, S.; Overbey, J.; Chen, K.; Lee, J.; Haj, D.; Li, Y. PD-L1’s Role in Preventing Alloreactive T Cell Responses Following Hematopoietic and Organ Transplant. Cells 2023, 12, 1609. https://doi.org/10.3390/cells12121609

AMA Style

Handelsman S, Overbey J, Chen K, Lee J, Haj D, Li Y. PD-L1’s Role in Preventing Alloreactive T Cell Responses Following Hematopoietic and Organ Transplant. Cells. 2023; 12(12):1609. https://doi.org/10.3390/cells12121609

Chicago/Turabian Style

Handelsman, Shane, Juliana Overbey, Kevin Chen, Justin Lee, Delour Haj, and Yong Li. 2023. "PD-L1’s Role in Preventing Alloreactive T Cell Responses Following Hematopoietic and Organ Transplant" Cells 12, no. 12: 1609. https://doi.org/10.3390/cells12121609

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

Article Metrics

Back to TopTop