Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many malignant and non-malignant hematological diseases. Donor T cells from the allografts are critical for the success of this effective therapy. Unfortunately these T cells not only recognize and attack the disease cells/tissues but also the other normal tissues of the recipient as ‘foreign’ or ‘nonself’ and cause severe immune mediated toxicity, Graft-versus-Host Disease (GVHD). Several insights into the complex pathophysiology of GVHD have been gained from recent experimental observations, which show that acute graft-vs. -host disease (GVHD) is a consequence of interactions between both the donor and the host innate and adaptive immune systems. These insights have identified a role for a variety of cytokines, chemokines, novel T cell subsets (naïve, memory, regulatory and NKT cells) and also for non-T cells of both the donor and host (antigen presenting cells, γδ T cells, B cells and and NK cells) in modulating the induction, severity and maintenance of acute GVHD. This review will focus on the immunobiology of experimental acute GVHD with an emphasis on the recent observations VSports最新版本.
Keywords: allogeneic, BMT, T cells, dendritic cells, antigens, cytokines
I. INTRODUCTION
Allogeneic hematopoietic cell transplantation (HCT) represents an important therapy for many hematological, some epithelial malignancies and also for a spectrum of non-malignant diseases(1). The development of novel strategies such as donor leukocyte infusions (DLI), nonmyeloablative HCT and cord blood transplantation (CBT) have helped expand the indications for allogeneic HCT over the last several years, especially among older patients(2). However, the major toxicity of allogeneic HCT, graft-versus-host disease (GVHD), remains a lethal complication that limits its wider application(3). Depending on the time at which it occurs after HCT, GVHD can be either acute or chronic(4–7). Acute GVHD is responsible for 15% to 40% of mortality and is the major cause of morbidity after allogeneic HCT, while chronic GVHD occurs up to 50% of patients who survive three months after HCT(1, 7). Research efforts over years have provided increasing insight into the biology of this complex disease process VSports注册入口.
The GVH reaction was first noted when irradiated mice were infused with allogeneic marrow and spleen cells(8). Although mice recovered from radiation injury and marrow aplasia, they subsequently died with ‘secondary disease(8). ’ a syndrome consisting of diarrhea weight loss, skin changes, and liver abnormalities. This phenomenon was subsequently recognized as GVH disease (GVHD). Three requirements for the development of GVHD were formulated by Billingham(9). First, the graft must contain immunologically competent cells, now recognized as mature T cells. In both experimental and clinical allogeneic BMT, the severity of GVHD correlates with the number of donor T cells transfused(10, 11). The precise nature of these cells and the mechanisms they use are now understood in greater detail (discussed below). Second, the recipient must be incapable of rejecting the transplanted cells i. e V体育官网入口. immuno-compromised. A patient with a normal immune system will usually reject cells from a foreign donor. In allogeneic BMT, the recipients are usually immuno-suppressed with chemotherapy and/or radiation before stem cell infusion(2). Third, the recipient must express tissue antigens that are not present in the transplant donor. This area has been the focus of intense research that has led to the discovery of the major histocompatibility complex (MHC)(12). Human Leukocyte Antigens (HLA) are proteins that are the gene products of the MHC and that are expressed on the cell surfaces of all nucleated cells in the human body. HLA proteins are essential to the activation of allogeneic T cells (12, 13) (discussed below).
This review on the pathophysiology of acute GVHD will place the genetic basis and the immuno-biological mechanisms of Billingham’s postulates in perspective.
II. GENETIC BASIS OF GVHD
Billingham’s third postulate stipulates that GVH reaction occurs when donor immune cells recognize disparate host antigens(9). These differences are governed by the genetic polymorphisms of the HLA system and the non-HLA systems such as the killer immunoglobulin receptors (KIR) family of NK receptors, nucleotide-binding oligomerization domain (NOD) 2 and cytokine gene polymorphisms(2, 13) V体育2025版.
A. HLA matching
Alloreactive T-cell-antigen recognition can be divided based on whether the presenting MHC molecule is matched or mismatched. For reasons as yet unclear, the precursor frequency of T cells that can recognize a mismatched MHC is very high (14–16). In humans, MHC is governed by the the HLA antigens that are encoded by the MHC gene complex on the short arm of chromosome 6 and can be categorized as Class I, II and III VSports. Class I antigens (HLA A, B, and C) are expressed on almost all cells of the body at varying densities (12). Class II antigens include DR, DQ, and DP antigens and are primarily expressed on hematopoietic cells (B cells, DCs, monocytes) although their expression can also be induced on many other cell types following inflammation or injury(12). The incidence of acute GVHD is directly related to the degree of MHC mismatch (17–20). The role of HLA mismatching in cord blood transplant (CBT) is more difficult to analyze than in unrelated HSCT, because allele level typing of CB units for HLA-A, B, C, DRB1, and DQB1 is not routinely performed(21). Nonetheless, the total number of HLA disparities between recipient and the CB unit has been shown to correlate with risk of acute GVHD and the frequency of severe acute GVHD was lower in patients transplanted with matched (6/6) CB units(21–23).
B. Minor histocompatibility antigens (MiHAs)
In MHC matched BMT context, as is the case with most clinical allo-BMT, donor T cells recognize MHC-bound peptides derived from the protein products of polymorphic genes (minor histocompatibility antigens or MiHAs) that are present in the host but not in the donor(24–28). Recent data suggest that genes not expressed in the donor but in the host can also generate relevant MiHAs(29, 30) VSports app下载.
Thus, despite HLA identity between a patient and donor, substantial numbers (40%) of patients receiving HLA-identical grafts and optimal post grafting immune suppression, develop acute GVHD due to differences in MiHAs that lie outside the HLA loci(27, 31). MiHAs are widely expressed, but can differ in their tissue expression(25, 31). This might be one of the reasons for the unique target organ involvement in GVHD. For example, most described human MiHAs although show wide but variable tissue expression pattern, but all are expressed in hematopoietic cells(25). This preponderance of MiHA expression on hematopoietic cells might account for making the host immune system a primary target for GVH response and further help explain the critical role of direct presentation by professional recipient antigen presenting cells (APCs) in causing anti-tumor and GVHD responses (by at least the donor CD8+ T cells) after allogeneic BMT. Experimental murine models have shown that different MiHAs dictate the phenotype, target organ involvement and the kinetics of GVHD(32). Recent efforts have identified some MiHAs such as, HA-1 and HA-2 that are primarily found on hematopoietic cells(33). These proteins may therefore induce GVHD. By contrast, other MiHAs, such as H-Y and HA-3, are expressed ubiquitously (see Table 1) (31) V体育官网. Experimental data have demonstrated that despite the presence of numerous potential MiHAs, all of the MiHAs are not equal in their ability to induce lethal GVHD and that they show hierarchical immunodominance(34, 35). Furthermore, difference in single immuno-dominant MiHAs alone was shown to be insufficient for causing GVHD in murine models although T cells targeting single MiHA can induce tissue damage in a skin explant model (36, 37). However, the role of specific and immuno-dominant MiHAs that are relevant in clinical GVHD has not been systematically evaluated in large groups of patients(38).
Table 1.
| Minor histocompatibility Antigens | Tissue Distribution |
|---|---|
| HA-1 | Hematopoietic |
| HA-2 | Hematopoietic |
| HB-1 | Hematopoietic |
| BCL2A1 | Hematopoietic |
| HA-3 | Ubiquitous |
| HA-8 | Ubiquitous |
| UGT2B17 | Ubiquitous |
| HY (A, B, DR, DQ) antigens | Ubiquitous |
V体育官网入口 - C. Other Non-HLA genes
Genetic polymorphisms in several non-HLA genes such as in KIRs, cytokines and NOD2 genes have recently been shown to modulate the severity and incidence of GVHD.
1. KIR polymorphisms
KIR receptors on NK cells that bind to the HLA class I gene products are encoded on chromosome 19. Polymorphisms in the trans-membrane and cytoplasmic domains of KIR receptors governs whether the receptor has inhibitory potential (such as KIR2DL1, -2DL2, -2DL3 and 3DL1 and their HLA class I ligands (HLA-C and HLA-Bw4) or activating potential; at this time, there is limited information on the clinical significance of activating KIR genes. Two competing models have been proposed for HLA-KIR allo-recognition by donor NK cells following HSCT: “mismatched ligand” and the “missing ligand” models(39–43). The former posits that NK alloreactivity occurs when donor NK cells recognize recipient target cells that lack the class I allele of the donor (HLA mismatching between the donor and recipient in the GVH direction); the latter hypothesizes that donor NK alloreactivity occurs when the host lacks the correct HLA class I ligand(s) to provide the inhibitory signal for donor KIR(42, 43). Both models are supported by some clinical observations, albeit in patients receiving very different transplant and immunosuppressive regimens(40, 44–46). Clearly, further validation is warranted and it is likely that the immuno-biology of the interface between HLA and KIR genetics will be an area of intense future investigation.
2. Cytokine gene polymorphisms
Pro-inflammatory cytokines, involved in the classical ‘cytokine storm’ of GVHD (discussed below), cause pathological damage of target organs such as skin, gut and liver(47). Several cytokine gene polymorphisms, both in hosts and donors, have been implicated. Specifically, TNF polymorphisms (TNFd3/d3 in the recipient, TNF-863 and -857 in donors and/or recipients and TNFd4, TNF-α-1031C and TNFRII-196R- in the donors) have been associated with an increased risk of acute GVHD and transplant-related mortality(48, 49). The three common haplotypes of the IL-10 gene promoter region in recipients representing high, intermediate and low production of IL-10 have been associated with the severity of acute GVHD following allo-BMT after HLA-matched sibling donors(50). By contrast, smaller studies have found neither IL-10 nor TNF-α polymorphisms to be associated with GVHD after HLA-mismatched cord blood transplants(49, 51). Furthermore, it is important to note that not all of the polymorphism analyses so far support the paradigm that excess of pro-inflammatory and Th1 cytokines are always associated with greater GVHD or mortality. For example IFN-γ polymorphisms of the 2/2 genotype (high IFN-γ production) and 3/3 genotype (low IFN-γ) have been associated with decreased or increased acute GVHD, respectively(49, 52). This is consistent with accelerated acute GVHD in IFN-γ knockout mouse models suggesting therefore that IFN-γ may be involved in the down-regulation of GVHD via a negative feed-back loop (discussed below)(53, 54). Similarly, possession of IL-6-174 polymorphism in the recipients (high production of IL-6) associated with both acute and chronic GVHD after MRD sibling HCT(52), which is in contrast to the results of IL-6 genetics in other Th1 mediated disease such as juvenile onset chronic arthritis (55).
By contrast, genotype (high production) of another Th1 cytokine, IL-2 (-330 allele G) was noted to increase the risk of acute GVHD in MUD transplants (56) while a different study showed that Th2 cytokine, IL-13, production by donor T cells is also predictive of acute GVHD in unrelated donor stem cell cohorts (57). Thus data from cytokine polymorphism studies so far do not seem to suggest a simple paradigm for GVHD. Nonetheless, the fact that most of the studies are small, not properly stratified must be considered while interpreting these data.
A small study in pediatric recipients of unrelated HSCT suggested that the presence of the IL-1α-889 allele in either donor or recipient decreased transplant related mortality but did not decrease GVHD(49). NOD2/CARD15 gene polymorphisms in both the donors and recipients were recently shown to have a striking association with GI GVHD and overall mortality after both related and unrelated allogeneic HSCT(58). It is likely non-HLA gene polymorphisms might play differing roles depending on the donor source (related vs. unrelated), HLA disparity (matched vs. mismatched), source of the graft (CB vs. PBSC vs. BM), and the intensity of the conditioning.
III. IMMUNOBIOLOGY
It is helpful to remember two important principles when considering the pathophysiology of acute GVHD. First, acute GVHD represents exaggerated but normal inflammatory responses against foreign antigens (allo-antigens) that are ubiquitously expressed in a setting where they are undesirable. The donor lymphocytes that have been infused into the recipient function appropriately, given the foreign environment they encounter. Second, donor lymphocytes encounter tissues in the recipient that have been often profoundly damaged. The effects of the underlying disease, prior infections, and the intensity of conditioning regimen all result in substantial changes not only in the immune cells but also in the endothelial and epithelial cells. Thus the allogeneic donor cells rapidly encounter not only a foreign environment, but one that has been altered to promote the activation and proliferation of inflammatory cells. Thus, the pathophysiology of acute GVHD may be considered a distortion of the normal inflammatory cellular responses that in addition to the absolute requirement of donor T cells, involves multiple other innate and adaptive cells and mediators(59). The development and evolution of acute GVHD can be conceptualized in three sequential phases (Figure 1) to provide a unified perspective on the complex cellular interactions and inflammatory cascades that lead to acute GVHD: (1) activation of the antigen presenting cells (APCs) (2) donor T cell activation, differentiation and migration and (3) effector phase(59).
Figure 1.

Three phases of GVHD immuno-biology
It is important to note that the three phase description as discussed below allows for a unified perspective in understanding the biology. It is, however, not meant to suggest that all three phases are of equal importance or that GVHD occurs in a step-wise and sequential manner. The spatio-temporal relationships between the biological processes described below, depending on the context, are more likely to be chaotic and of varying intensity and relevance in the induction, severity and maintenance of GVHD.
A. Phase 1: Activation of antigen presenting cells (APCs)
The earliest phase of acute GVHD is set into motion by the profound damage caused by the underlying disease and infections and further exacerbated by the BMT conditioning regimens (which include total body irradiation (TBI) and/or chemotherapy) that are administered even before the infusion of donor cells(60–64). This first step results in activation of the APCs. Specifically, damaged host tissues respond with multiple changes, including the secretion of proinflammatory cytokines, such as TNF-α and IL-1, described as the ‘cytokine storm’(62, 63, 65). Such changes increase expression of adhesion molecules, costimulatory molecules, MHC antigens and chemokines gradients that alert the residual host and the infused donor immune cells(63). These “danger signals” activate host APCs(66, 67). Damage to the gastrointestinal (GI) tract from the conditioning is particularly important in this process because it allows for systemic translocation of immuno-stimulatory microbial products such as lipopolysaccaride (LPS) that further enhance the activation of host APCs and the secondary lymphoid tissue in the GI tract is likely the initial site of interaction between activated APCs and donor T cells (63) (68, 69). This scenario accords with the observation that an increased risk of GVHD is associated with intensive conditioning regimens that cause extensive injury to epithelial and endothelial surfaces with a subsequent release of inflammatory cytokines and increases in expression of cell surface adhesion molecules(63, 64). The relationship among conditioning intensity, inflammatory cytokine, and GVHD severity has been supported by elegant murine studies(65). Furthermore, the observations from these experimental studies have led to two recent clinical innovations to reduce clinical acute GVHD: (a) reduced intensity conditioning to decrease the damage to host tissues and thus limit activation of host APC and (b) KIR mismatches between donor and recipients to eliminate the host APCs by the alloreactive NK cells(41, 70).
Host type APCs that are present and have been primed by conditioning, are critical for the induction of this phase; recent evidence suggests that donor type APCs exacerbate GVHD, but, in certain experimental models, donor type APC chimeras also induce GVHD(67, 71–73). In clinical situations, if donor type APCs are present in sufficient quantity and have been appropriately primed, they too might play a role in the initiation and exacerbation of GVHD(74–76). Amongst the cells with antigen presenting capability, DCs are the most potent and play an important role in the induction of GVHD(77). Experimental data suggest that GVHD can be regulated by qualitatively or quantitatively modulating distinct DC subsets(78–83). Langerhans cells were also shown to be sufficient for the induction of GVHD when all other APCs were unable to prime donor T cells, although the role for Langerhans cells when all APCs are intact is unknown(84). Studies have yet to define roles for other DC subsets. In one clinical study persistence of host DC after day 100 correlated with the severity of acute GVHD while elimination of host DCs was associated with reduced severity of acute GVHD(75). The allo-stimulatory capacity of mature monocyte derived DCs (mDCs) after reduced intensity transplants was lower for up to six months compared to the mDCs from myeloablative transplant recipients, thus suggesting a role for host DCs and the reduction in ‘danger signals’ secondary to less intense conditioning in acute GVHD(85). Nonetheless this concept of enhanced host APC activation explains a number of clinical observations such as increased risks of acute GVHD associated with advanced stage malignancy, conditioning intensity and histories of viral infections.
Other professional APCs such as monocytes/macrophages or semi-professional APCs might also play a role in this phase. For example, recent data suggests that host type B cells might play a regulatory role under certain contexts(86). Also host or donor type non-hematopoietic stem cells, such as mesenchymal stem cells or stromal cells when acting as APCs have been shown to reduce T cell allogeneic responses, although the mechanism for such inhibition remains unclear. The relative contributions of various APCs, professional or otherwise, remain to be elucidated.
B. Phase 2: Donor-T-Cell Activation, differentiation and migration
The infused donor T cells interact with the primed APCs leading to the initiation of the second phase of acute GVHD. This phase includes antigen presentation by primed APCs, the subsequent activation, proliferation, differentiation and migration of alloreactive donor T cells.
After allogeneic HSC transplants, both host- and donor-derived APCs are present in secondary lymphoid organs(87, 88). The T-cell receptor (TCR) of the donor T cells can recognize alloantigens either on host APCs (direct presentation) or donor APCs (indirect presentation)(89, 90). In direct presentation, donor T cells recognize either the peptide bound to allogeneic MHC molecules or allogeneic MHC molecules without peptide (90, 91). During indirect presentation, T cells respond to the peptide generated by degradation of the allogeneic MHC molecules presented on self-MHC (91). Experimental study demonstrated that APCs derived from the host, rather than from the donor, are critical in inducing GVHD across MiHA mismatch (89). Recent data suggest that presentation of distinct target antigens by the host and donor type APCs might play a differential role in mediating target organ damage(32, 92). In humans, most cases of acute GVHD developed when both host DCs and donor dendritic cells (DCs) are present in peripheral blood after BMT (75).
1. Co-stimulation
The interaction of donor lymphocyte TCR with the host allo-peptide presented on the MHC of APCs alone is insufficient to induce T cell activation(93). Both TCR ligation and co-stimulation via a ‘second’ signal through interaction between the T cell co-stimulatory molecules and their ligands on APCs are required to achieve T proliferation, differentiation and survival(94). The danger signals generated in phase 1 augment these interactions and significant progress has been made on the nature and impact of these ‘second’ signals(95, 96). Costimulatory pathways are now known to deliver both positive and negative signals and molecules from two major families, the B7 family and the TNF receptor (TNFR) family play pivotal roles in GVHD(97). Interruption of the second signal by blockade of various positive co-stimulatory molecules (CD28, ICOS, CD40, CD30, 4-1BB and OX40) reduces acute GVHD in several murine models while antagonism of the inhibitory signals (PD-1 and CTLA-4) exacerbates the severity of acute GVHD(2, 98–103). The various T cell and APC co-stimulatory molecules and the impact on acute GVHD are summarized in Table 2. The specific context and the hierarchy in which each of these signals play a dominant role in the modulation of GVHD remain to be determined.
Table 2.
T Cell co-stimulation
| T cell | APC | |
|---|---|---|
| Adhesion | ICAMs | LEA-I |
| LEA-1 | ICAM~ | |
| CD2 (LEA-2) | LFA-3 | |
| Recognition | TCR/CD4 | NIIIC hi |
| TCR/CD8 | Mi-Icc I | |
| Costimulation | CD28 | CD80/86 |
| CD152 (CTLA-4 | CD8O/86 | |
| ICOS | B7H/B7RP-1 | |
| PD-1 | PD-L1, PD-L2 | |
| Unknown | B7-H3 | |
| CD 154 (CD4OL) | CD4O | |
| CD134 (0X40) | CD134L (OX4OL) | |
| CD137 (4-IBB) | CDI37L (4-1IBBL) | |
| HVEM | LIGHT |
Abbreviations: HVEM HSV glycoprotein D for herpesvirus entry mediator; LIGHT, homologous to lymphotoxins, shows inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator (HVEM), a receptor expressed by T lymphocytes.
2. T cell subsets
T cells consist of several subsets whose responses differ based on antigenic stimuli, activation thresholds and effector functions. The alloantigen composition of the host determines which donor T-cell subsets proliferate and differentiate.
CD4+ and CD8+ cells
CD4 and CD8 proteins are co-receptors for constant portions of MHC class II and class I molecules, respectively(104). Therefore MHC class I (HLA-A, -B, -C) differences stimulate CD8+Tcells and MHC class II (HLA-DR -DP, -DQ) differences stilnulateCD4+T cells(104–107). But clinical trials of CD4+ or CD8+ depletion have been inconclusive(108). This perhaps is not surprising, because GVHD is induced by MiHAs in the majority of HLA-identical BMT, which are peptides derived from polymorphic cellular proteins that are presented by MHC molecules(28). Because the manner of protein processing depends on genes of the MHC, two siblings will have many different peptides in the MHC groove(28). Thus in the majority of HLA-identical BMT, acute GVHD may be induced by either or both CD4+ and CD8+ subsets in response to minor histocompatibility antigens(108). The peptide repertoire for class I or class II MHC remains unknown and might even be different in different individuals(109). But it is plausible that only a few of the many of these peptides might behave as immuno-dominant “major minor” antigens that can potentially induce GVHD. In any event, such antigens remain to be identified and validated in large patient population.
Central deletion by establishment of stable mixed hematopoietic chimeric state is an effective way to eliminate continued thymic production of both CD4+ and CD8+ alloreactive T cells and thus reduce GVHD(110–112). In contrast peripheral mechanisms to induce tolerance of CD4+ and CD8+ T cells appears to be distinct(113, 114). The pathways of T cell apoptosis by which peripheral deletion occurs can be broadly categorized into activation-induced cell death (AICD) and passive cell death (PCD)(115). Experimental data suggests that deletional tolerance by AICD is operative via the Fas (for CD4+) or TNFR (CD8+) pathways in Th1 cells and when the frequency of alloreactive T cells is at much greater(116–121). PCD or ‘death by neglect’ is due to rapid down regulation of Bcl-2 and appears to be critical in non-irradiated but not after irradiated BMT(122). Thus distinct mechanisms of tolerance induced by apoptosis have a dominant role depending on the T cell subsets, the conditioning regimens and the histocompatibility differences. Nonetheless strategies aimed at selective elimination of donor T cells in vivo after HCT either by targeting a suicide gene to the allo-T cells or by photodynamic cell purging appears to be promising in reducing experimental acute GVHD(123–129).
Naïve and Memory subsets
Several independent groups have intriguingly found that the naïve (CD62L+) T cells were alloreactive and caused acute GVHD but not the memory (CD62L−) T cells across different donor/recipient strain combinations(130–133). Furthermore, expression of naïve T cell marker CD62L was also found to be critical for regulation of GVHD by donor natural regulatory T cells(134, 135). By contrast, another recent study demonstrated that alloreactive memory T cells and their precursor cells (memory stem cells) caused robust GVHD(136, 137). It remains as yet unknown whether the reduced GVHD potential of memory type T cells from a naïve murine donor, in contrast to their ability to cause greater solid organ allo-rejection(138), is due to a consequence of the intense conditioning regimen and/or altered trafficking or from a restricted repertoire and/or from T cell intrinsic defect.
Regulatory T cells
Recent advances indicate that distinct subsets of regulatory CD4+CD25+, CD4+CD25−IL10+ Tr cells, γδT cells, DN− T cells, NK T cells and regulatory DCs control immune responses by induction of anergy or active suppression of alloreactive T cells(79, 80, 139–147). Several studies have demonstrated a critical role for the natural donor CD4+CD25+ Foxp3+ regulatory T (Treg) cells, obtained from naïve animals or generated ex-vivo, in the outcome of acute GVHD. Donor CD4+CD25+ T cells suppressed the early expansion of alloreactive donor T cells and their capacity to induce acute GVHD without abrogating GVL effector function against these tumors(148, 149). CD4+CD25+ T cells induced/generated by of immature or regulatory host type DCs and by regulatory donor type myeloid APCs were also able to suppress acute GVHD(79). One of the clinical studies that evaluated the relationship between donor CD4+CD25+ cells and acute GVHD in humans after matched sibling donor grafts and found that in contrast to the murine studies, donor grafts containing larger numbers of CD4+ CD25+T cells developed more severe acute GVHD(150). These data suggest that co-expression of CD4+ and CD25+ is insufficient because an increase in CD25+ T cells in donor grafts is associated with greater risks of acute GVHD after clinical HCT. Another recent study found that Foxp3 mRNA expression (considered a specific marker for naturally occurring CD4+CD25+Tregs) was significantly decreased in peripheral blood mononuclear cells from patients with acute GVHD(151, 152). But Foxp3 expression in humans, unlike mice, may not be specific for T cells with a regulatory phenotype(153). It is likely that the precise role of regulatory T cells in clinical acute GVHD will therefore not only depend upon identification of specific molecular markers in addition to Foxp3 but also on the ability for ex vivo expansion of these cells in sufficient numbers. Several clinical trials are underway in the US and Europe with attempts to substantially expand these cells ex-vivo and use for prevention of GVHD.
Host NK1.1+ T cells are another T cell subsets with suppressive functions have also been shown to suppress acute GVHD in an IL-4 dependent manner(146, 147, 154). By contrast, donor NKT cells were found to reduce GVHD and enhance perforin mediated GVL in an IFN-γ dependent manner(155, 156). Recent clinical data suggests that enhancing recipient NKT cells by repeated TLI conditioning promoted Th2 polarization and dramatically reduced GVHD(147). Experimental data also show that activated donor NK cells can reduce GVHD through the elimination of host APCs or by secretion of transforming growth factor-β (TGF-β) secretion(156). A murine BMT study using mice lacking SH2-containing inositol phosphatase (SHIP), in which the NK compartment is dominated by cells that express two inhibitory receptors capable of binding either self or allogeneic MHC ligands, suggests that host NK cells may play a role in the initiation of GVHD(157).
3. Cytokines and T cell differentiation
APC and T cell activation result in rapid intracellular biochemical cascades that induce transcription of many genes including cytokines and their receptors. The Th1 cytokines (IFN-γ, IL-2 and TNF-α) have been implicated in the pathophysiology of acute GVHD(158–160). IL-2 production by donor T cells remains the main target of many current clinical therapeutic and prophylactic approaches, such as cyclosporine, tacrolimus and monoclonal antibodies (mAbs) against the IL-2 and its receptor to control acute GVHD(161, 162). But emerging data indicate an important role for IL-2 in the generation and maintenance of CD4+CD25+ Foxp3+ T regs, suggesting that prolonged interference with IL-2 may have an unintended consequence in the prevention of the development of long term tolerance after allogeneic HCT(163–166).
Studies by Sykes and colleagues and others have demonstrated that the role of Th1 cytokines is complex. For example exogenous administration of IFN-γ or T cells from IFN-γ deficient donors have demonstrated a reduction and enhancement of GVHD respectively(53, 54, 167, 168). These data are consistent with the clinical IFN-γ polymorphism data (discussed above). A recent study suggests that IFN- γ might play a differential role in the severity of distinct GVHD target organs(169). Likewise, early injection of IL-2 has also been shown to reduce GVHD(170, 171). Thus whether the Th1 cytokines are the regulators or inducers of GVHD severity depends on the degree of allo-mismatch, the intensity of conditioning and the T cell subsets that are involved after BMT(172–174). Thus although the “cytokine storm” initiated in phase 1 and amplified by the Th1 cytokines correlates with the development of acute GVHD, early Th1 polarization of donor T cells to HCT recipients can attenuate acute GVHD suggesting that physiological and adequate amounts of Th1 cytokines are critical for GVHD induction, while inadequate production (extremely low or high) could modulate acute GVHD through a breakdown of negative feedback mechanisms for activated donor T cells(160, 174–177). Several different cytokines that polarize donor T cells to Th2 such as IL-4, G-CSF, IL-18, IL-11, rapamycin and the secretion of IL-4 by NK1.1+ T cells can reduce acute GVHD(178–185). But Th1 and Th2 subsets cause injury of distinct acute GVHD target tissues and some studies failed to show a beneficial effect of Th2 polarization on acute GVHD(186). Thus the Th1/Th2 paradigm of donor T cells in the immuno-pathogenesis of acute GVHD has evolved over the last few years and its causal role in acute GVHD is complex and incompletely understood.
IL-10 plays a key role in suppression of immune responses and its role in regulating experimental acute GVHD is unclear(187). Recent clinical data demonstrate an unequivocal association of IL-10 polymorphisms with the severity of acute GVHD(50). TGF-β, another suppressive cytokine was shown to suppress acute GVHD but to exacerbate chronic GVHD(188). The roles of some other cytokines, such as IL-7 (that promotes immune reconstitution) and IL-13 remain unclear(189–192). The role for Th17 cells, a recently described novel T cell differentiation in many immunological processes, is not yet known(193). In any case, all of the experimental data so far collectively suggest that the timing of administration, the production of any given cytokine, the intensity of the conditioning regimen and the donor-recipient combination may all be critical to the eventual outcome of acute GVHD.
4. Leukocyte migration
Donor T cells migrate to lymphoid tissues, recognize alloantigens on either host or donor APCs and become activated. They then exit the lymphoid tissues and traffic to the target organs and cause tissue damage(194). The molecular interactions necessary for T cell migration and the role of lymphoid organs during acute GVHD have recently become the focus of a growing body of research. Chemokines play a critical role in the migration of immune cells to secondary lymphoid organs and target tissues(195). T-lymphocyte production of macrophage inflammatory protein-1alpha (MIP-1α) is critical to the recruitment of CD8+ but not CD4+ T cells cells to the liver, lung, and spleen during acute GVHD(196). Several chemokines such as CCL2-5, CXCL2, CXCL9-11, CCL17 and CCL27 are over-expressed and might play a critical role in the migration of leukocyte subsets to target organs liver, spleen, skin and lungs during acute GVHD(194, 197). CXCR3+ T and CCR5+ T cells cause acute GVHD in the liver and intestine(194, 198–200). CCR5 expression has also been found to be critical for Treg migration in GVHD(201). In addition to chemokines and their receptors, expression of selectins and integrins and their ligands also regulate the migration of inflammatory cells to target organs(195). For example, interaction between α4β7 integrin and its ligand MadCAM-1 are important for homing of donor T cells to Peyer’s patches and in the initiation of intestinal GVHD(68, 202). αLβ2/ICAM1, 2, 3 and α4β1/VCAM-2 interactions are important for homing to the lung and liver after experimental HCT(194). The expression of CD62L on donor Tregs is critical for their regulation of acute GVHD suggesting that their migration in secondary tissues is critical for their regulatory effects(88). The migratory requirement of donor T cells to specific lymph nodes (e.g. Peyer’s patches) for the induction of GVHD might depend on other factors such as the conditioning regimen, inflammatory milieu etc(68, 203). Furthermore, FTY720, a pharmacologic sphingosine-1-phosphate receptor agonist, inhibited GVHD in murine but not in canine models of HCT(204, 205). Thus, there might also be significant species differences in the ability of these molecules to regulate GVHD.
C. Phase 3: Effector Phase
The effector phase that leads to the GVHD target organ damage is a complex cascade of multiple cellular and inflammatory effectors that further modulate each others responses either simultaneously or successively. Effector mechanisms of acute GVHD can be grouped into cellular effectors (e.g., CTLs) and inflammatory effectors such as cytokines. Inflammatory chemokines expressed in inflamed tissues upon stimulation by proinflammatory effectors such as cytokines are specialized for the recruitment of effector cells, such as CTLs(206). Furthermore the spatio-temporal expression of the cyto-chemokine gradients might determine not only the severity but also the unusual cluster of GVHD target organs (skin, gut, and liver)(194, 207).
1. Cellular Effectors
Cytotoxic T cells (CTLs) are the major cellular effectors of GVHD(208, 209). The Fas-Fas ligand (FasL), the perforin-grazyme (or granule exocytosis) and TNFR-like death receptors (DR), such as TNF-related apoptosis-inducing ligand (TRAIL: DR4, 5 ligand) and TNF-like weak inducers of apoptosis (TWEAK: DR3 ligand) are the principle CTL effector pathways that have been evaluated after allogeneic BMT(209–214). The involvement of each of these molecules in GVHD has been testing by utilizing donor cells that are unable to mediate each pathway. Perforin is stored in cytotoxic granules of CTLs and NK cells, together with granzymes and other proteins. Although the exact mechanisms remain unclear, following the recognition of a target cell through the TCR-MHC interaction, perforin is secreted and inserted into the cell-membrane, forming “perforin pores” that allow granzymes to enter the target cells and induce apoptosis through various downstream effector pathways such as caspases(215). Ligation of Fas results in the formation of the death-inducing signaling complex (DISC) and also activates caspases(216, 217).
Transplantation of perforin deficient T cells results in a marked delay in the onset of GVHD in transplants across MiHA disparities only, both MHC and MiHA disparities (126), and across isolated MHC I or II disparities(209, 218–222). However, mortality and clinical and histological signs of GVHD were still induced even in the absence of perforin-dependent killing in these studies, demonstrating that the perforin-granzyme pathways plays little role in target organ damage. A role for the perforin-granzyme pathway for GVHD induction is also evident in studies employing donor-T-cell subsets. Perforin- or granzyme B-deficient CD8+ T cells caused less mortality than wild-type T cells in experimental transplants across a single MHC class I mismatch. This pathway, however, seems to be less important compared to Fas/FasL pathway in CD4-mediated GVHD(221–223). Thus, it seems that CD4+ CTLs preferentially use the Fas-FasL pathway, whereas CD8+CTLs primarily use the perforin-granzyme pathway.
Fas, a TNF-receptor family member, is expressed by many tissues, including GVHD target organs(224). Its expression can be upregulated by inflammatory cytokines such as IFN-γ and TNF-α during GVHD, and the expression of FasL is also increased on donor T cells, indicating that FasL-mediated cytotoxicity may e a particularly important effector pathway in GVHD(209, 225). FasL-defective T cells cause less GVHD in the liver, skin and lymphoid organs(220, 223, 225). The Fas-FasL pathway is particularly important in hepatic GVHD, consistent with the keen sensitivity of hepatocytes to Fas-mediated cytotoxicity in experimental models of murine hepatitis(209). Fas-deficient recipients are protected from hepatic GVHD, but not from other organ GVHD, and administration of anti-FasL (but not anti-TNF) MAbs significantly blocked hepatic GVHD damage occurring in murine models(209, 226, 227). Although the use of FasL-deficient donor T cells or the administration of neutralizing FasL MAbs had no effect on the development of intestinal GVHD in several studies, the Fas-FasL pathway may play a role in this target organ, because intestinal epithelial lymphocytes exhibit increased FasL-mediated killing potential(228). Elevated serum levels of soluble FasL and Fas have also been observed in at least some patients with acute GVHD(229, 230).
The utilization of a perforin-granzyme and FasL cytotoxic double-deficient (cdd) mouse provides an opportunity to address whether other effector pathways are capable of inducing GVHD target organ pathology. An initial study demonstrated that cdd T cells were unable to induce lethal GVHD across MHC class I and class II disparities after sublethal irradiation(219). However, subsequent studies demonstrated that cytotoxic effector mechanisms of donor T ells are critical in preventing host resistance to GVHD(213, 231). Thus when recipients were conditioned with lethal dose of irradiation, cdd CD4+ T cells produced similar mortality to wild type CD4+ T cells(213). These results were confirmed by a recent study demonstrating that GVHD target damage can occur in mice that lack alloantigen expression on the epithelium, preventing direct interaction between CTLs and target cells(214).
The participation of another death ligand receptor signaling pathway, TNF/TNFRs, has also been evaluated. Experimental data suggests that this pathway is crucial for GI GVHD (discussed more below). Recently, several additional TNF family apoptosis-inducing receptors/ligands have been identified, including TWEAK, TRAIL and LTβ/LIGHT have all been proposed to play a role in GVHD and GVL responses(2, 232–238). However whether these distinct pathways play a more specific role for GVHD mediated by distinct T cell subsets in certain situations remains unknown. Intriguingly, recent data suggest that none of these pathways might be critical for mediating the rejection of donor grafts(232, 239). Thus it is likely that their role in GVHD might be modulated by the intensity of conditioning and by the recipient T cell subsets. Existing experimental data suggest that perforin and TRAIL cytotoxic pathways are associated with CD8+ T cell–mediated GVL(209). The available experimental data are strongly skewed toward CD8+ T cell–mediated GVL based on the dominant role of this effector population in most murine GVT models; however, CD4+ T cells can mediate GVL and might be crucial in clinical BMT depending on the type of malignancy and the expression of immuno-dominant antigens.
Taken together, although experimental data suggest that might be some distinction between the use of different lytic pathways for the specific GVHD target organs and GVL, but the clinical applicability of these observations is as yet largely unknown
2. Inflammatory Effectors
Inflammatory cytokines synergize with CTLs resulting in the amplification of local tissue injury and further promotion of an inflammation, which ultimately leads to the observed target tissue destruction in the transplant recipient(47). Macrophages, which had been primed with IFN-γ during step 2, produce inflammatory cytokines TNF-α and IL-1 when stimulated by a secondary triggering signal(240). This stimulus may be provided through Toll-like receptors (TLRs) by microbial products such as LPS and other microbial particles, which can leak through the intestinal mucosa damaged by the conditioning regimen and gut GVHD(241, 242). It is now apparent that immune recognition through both TLR and non-TLRs (such as NOD) by the innate immune system also controls activation of adaptive immune responses(241, 243). Recent clinical studies of GVHD suggested the possible association with TLR/NOD polymorphisms and severity of GVHD(58, 244, 245). LPS and other innate stimuli may stimulate gut-associated lymphocytes, keratinocytes, dermal fibroblasts, and macrophages to produce pro-inflammatory effectors that play a direct role in causing target organ damage. Indeed experimental data with MHC mismatched BMT suggest that under certain circumstances these inflammatory mediators are sufficient in causing GVHD damage even in the absence of direct CTL induced damage(71). The severity of GVHD appears to be directly related to the level of innate and adaptive immune cell priming and release of pro-inflammatory cytokines such as TNF- α, IL-1 and nitric oxide (NO)(71, 242, 246–248).
The cytokines TNF- α and IL-1 are produced by an abundance of cell types during processes of both innate and adaptive immunity; they often have synergistic, pleiotrophic, and redundant effects on both activation and effector phases of GVHD(160). A critical role for TNF- α in the pathophysiology of acute GVHD was first suggested over 20 years ago because mice transplanted with mixtures of allogeneic BM and T cells developed severe skin, gut, and lung lesions that were associated with high levels of TNF- α mRNA in these tissues(249). Target organ damage could be inhibited by infusion of anti-TNF- α MAbs, and mortality could be reduced from 100% to 50% by the administration of the soluble form of the TNF- α receptor (sTNFR), an antagonist of TNF- α(62, 65, 247). Accumulating experimental data further suggest that TNF -α is involved in a multistep process of GVHD pathophysiology. TNF-α can (1) cause cachexia, a characteristic feature of GVHD, (2) induce maturation of DCs, thus enhancing alloantigen presentation, (3) recruit effector T cells, neutrophilis, and monocytes into target organs through the induction of inflammatory chemokines, and (4) cause direct tissue damage by inducing apoptosis and necrosis. TNF-α also involves in donor-T-cell activation directly trough it’s signaling via TNFR1 and TNFR2 on T cells. TNF-TNF1 interactions on donor T cells promote alloreactive T-cell responses and TNF-TNFR2 interactions are critical for intestinal GVHD(235, 250). TNF-α also seems to be important effector molecules in GVHD in skin and lymphoid tissue(249, 251). Additionally, TNF-α might also be involved in hepatic GVHD, probably by enhancing effector cell migration to the liver via the induction of inflammatory chemokines(252). An important role for TNF-α in clinical acute GVHD has been suggested by studies demonstrating elevated serum levels or TNF- α or elevated TNF- α mRNA expression in peripheral blood mononuclear cells in patients with acute GVHD and other endothelial complications, such as hepatic veno-occlusive disease (VOD)(252–255). Phase I–II trials using TNF-α antagonists reduced the severity of GVHD suggesting that it is a relevant effector in causing target organ damage(256, 257).
The second major pro-inflammatory cytokine that appears to play an important role in the effector phase of acute GVHD is IL-1(258). Secretion of IL-1 appears to occur predominantly during the effector phase of GVHD of the spleen and skin, two major GVHD target organs(259). A similar increase in mononuclear cell IL-1 mRNA has been shown during clinical acute GVHD. Indirect evidence of a role for IL-1 in GVHD was obtained with administration of this cytokine to recipients in an allogeneic murine BMT model. Mice receiving IL-1 displayed a wasting syndrome and increased mortality that appeared to be an accelerated form of disease. By contrast, intra-peritoneal administration of IL-1ra starting on d 10 post-transplant was able to reverse the development of GVHD in the majority of animals, providing a significant survival advantage to treated animals(260). However, the attempt to use IL-1ra to prevent acute GBHD in a randomized trial was not successful(261).
As a result of activation during GVHD, macrophages also produce NO, which contributes to the deleterious effects on GVHD target tissues, particularly immunosuppression(248, 262). NO also inhibits the repair mechanisms of target tissue destruction by inhibiting proliferation of epithelial stem cells in the gut and skin(263). In humans and rats, the development of GVHD is preceded by an increase in serum levels of NO oxidation products(264–267).
Existing data demonstrate important role for various inflammatory effectors in GVHD. The relevance of currently studied or as yet unknown specific effectors might however be determined by other factors, including the intensity of preparatory regimens, the type of allograft, the T cell subsets and the duration of BMT. In any event, both experimental and clinical data suggest an important role for both the cellular and inflammatory mediators in GVHD induced target organ damage.
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 citable 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.Appelbaum FR. Haematopoietic cell transplantation as immunotherapy. Nature. 2001;411:385–9. doi: 10.1038/35077251. [DOI] [PubMed] [Google Scholar]
- 2.Welniak LA, Blazar BR, Murphy WJ. Immunobiology of Allogeneic Hematopoietic Stem Cell Transplantation. Annu Rev Immunol. 2006 doi: 10.1146/annurev.immunol.25.022106.141606. [V体育2025版 - DOI] [PubMed] [Google Scholar]
- 3.Ferrara JL, Reddy P. Pathophysiology of graft-versus-host disease. Semin Hematol. 2006;43:3–10. doi: 10.1053/j.seminhematol.2005.09.001. [DOI] [PubMed] [Google Scholar]
- 4.Deeg HJ. How I Treat Refractory Acute GVHD. Blood. 2007 doi: 10.1182/blood-2006-12-041889. [DOI (VSports手机版)] [PMC free article] [PubMed] [Google Scholar]
- 5.Weiden PL, Flournoy N, Thomas ED, Prentice R, Fefer A, Buckner CD, Storb R. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979;300:1068–73. doi: 10.1056/NEJM197905103001902. ["V体育官网入口" DOI] [PubMed] [Google Scholar]
- 6.Weiden PL, Sullivan KM, Flournoy N, Storb R, Thomas ED. Antileukemic effect of chronic graft-versus-host disease: Contribution to improved survival after allogeneic marrow transplantation. N Engl J Med. 1981;304:1529–33. doi: 10.1056/NEJM198106183042507. [V体育2025版 - DOI] [PubMed] [Google Scholar]
- 7.Lee SJ. New approaches for preventing and treating chronic graft-versus-host disease. Blood. 2005;105:4200–6. doi: 10.1182/blood-2004-10-4023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.van Bekkum DW, De Vries MJ. Radiation chimaeras. London: Logos Press; 1967. [Google Scholar]
- 9.Billingham RE. The biology of graft-versus-host reactions. Harvey Lec. 1966;62:21–78. [PubMed] [Google Scholar]
- 10.Korngold R, Sprent J. Purified T cell subsets and lethal graft-versus-host disease in mice. In: Gale RP, Champlin R, editors. Progress in Bone Marrow Transplantation. New York: Alan R. Liss, Inc; 1987. pp. 213–8. [Google Scholar]
- 11.Kernan NA, Collins NH, Juliano L, Cartagena T, Dupont B, RJ OR. Clonable T lymphocytes in T cell-depleted bone marrow transplants correlate with development of graft-v-host disease. Blood. 1986;68:770–3. [PubMed] [Google Scholar]
- 12.Petersdorf EW, Malkki M. Genetics of risk factors for graft-versus-host disease. Semin Hematol. 2006;43:11–23. doi: 10.1053/j.seminhematol.2005.09.002. ["V体育平台登录" DOI] [PubMed] [Google Scholar]
- 13.Krensky AM, Weiss A, Crabtree G, Davis MM, Parham P. T-lymphocyte-antigen interactions in transplant rejection. N Engl J Med. 1990;322:510–7. doi: 10.1056/NEJM199002223220805. [DOI] [PubMed] [Google Scholar]
- 14.Aosai F, Ohlen C, Ljunggren HG, Hoglund P, Franksson L, Ploegh H, Townsend A, Karre K, Stauss HJ. Different types of allospecific CTL clones identified by their ability to recognize peptide loading-defective target cells. Eur J Immunol. 1991;21:2767–74. doi: 10.1002/eji.1830211118. [DOI] [PubMed] [Google Scholar]
- 15.Wang W, Man S, Gulden PH, Hunt DF, Engelhard VH. Class I-restricted alloreactive cytotoxic T lymphocytes recognize a complex array of specific MHC-associated peptides. J Immunol. 1998;160:1091–7. [PubMed] [Google Scholar]
- 16.Man S, Salter RD, Engelhard VH. Role of endogenous peptide in human alloreactive cytotoxic T cell responses. Int Immunol. 1992;4:367–75. doi: 10.1093/intimm/4.3.367. [DOI (V体育ios版)] [PubMed] [Google Scholar]
- 17.Anasetti C, Amos D, Beatty PG, Appelbaum FR, Bensinger W, Buckner CD, Clift R, Doney K, Martin PJ, Mickelson E, et al. Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma. N Engl J Med. 1989;320:197–204. doi: 10.1056/NEJM198901263200401. [DOI (V体育官网入口)] [PubMed] [Google Scholar]
- 18.Petersdorf EW, Longton G, Anasetti C, et al. Donor-recipient disparities for HLA-C genes is a risk factor for graft failure following marrow transplantation from unrelated donors. Blood 86, Suppl. 1995;1:291a. [Google Scholar]
- 19.Petersdorf EW, Longton GM, Anasetti C, Mickelson EM, McKinney SK. Association of HLA-C Disparity with Great Failure After Marrow Transplantation from Unrelated Donors. Blood. 1997;89:1818–23. [VSports - PubMed] [Google Scholar]
- 20.Flomenberg N, Baxter-Lowe LA, Confer D, Fernandez-Vina M, Filipovich A, Horowitz M, Hurley C, Kollman C, Anasetti C, Noreen H, Begovich A, Hildebrand W, Petersdorf E, Schmeckpeper B, Setterholm M, Trachtenberg E, Williams T, Yunis E, Weisdorf D. Impact of HLA class I and class II high-resolution matching on outcomes of unrelated donor bone marrow transplantation: HLA-C mismatching is associated with a strong adverse effect on transplantation outcome. Blood. 2004;104:1923–30. doi: 10.1182/blood-2004-03-0803. [DOI] [PubMed] [Google Scholar]
- 21.Barker JN, Wagner JE. Umbilical-cord blood transplantation for the treatment of cancer. Nat Rev Cancer. 2003;3:526–32. doi: 10.1038/nrc1125. [DOI] [PubMed] [Google Scholar]
- 22.Laughlin MJ, Eapen M, Rubinstein P, Wagner JE, Zhang MJ, Champlin RE, Stevens C, Barker JN, Gale RP, Lazarus HM, Marks DI, van Rood JJ, Scaradavou A, Horowitz MM. Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia. N Engl J Med. 2004;351:2265–75. doi: 10.1056/NEJMoa041276. [DOI] [PubMed] [Google Scholar]
- 23.Barker JN, Weisdorf DJ, DeFor TE, Blazar BR, McGlave PB, Miller JS, Verfaillie CM, Wagner JE. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005;105:1343–7. doi: 10.1182/blood-2004-07-2717. [DOI] [PubMed] [Google Scholar]
- 24.Den Haan JM, Sherman NE, Blokland E, Huczko E, Koning F, Drijfhout JW, Skipper J, Shabanowitz J, Hunt DF, Engelhard VH, et al. Identification of a graft versus host disease-associated human minor histocompatibility antigen. Science. 1995;268:1476–80. doi: 10.1126/science.7539551. [DOI] [PubMed] [Google Scholar]
- 25.de Bueger M, Bakker A, Van Rood JJ, Van der Woude F, Goulmy E. Tissue distribution of human minor histocompatibility antigens. Ubiquitous versus restricted tissue distribution indicates heterogeneity among human cytotoxic T lymphocyte-defined non-MHC antigens. J Immunol. 1992;149:1788–94. [PubMed] [Google Scholar]
- 26.de Bueger M, Goulmy E. Human minor histocompatibility antigens. Transpl Immunol. 1993;1:28–38. doi: 10.1016/0966-3274(93)90056-e. [DOI] [PubMed] [Google Scholar]
- 27.Goulmy E, Schipper R, Pool J, Blokland E, Falkenburg F. Mismatches of Minor Histocompatibility Antigens Between HLA-Identical Donors and Recipients and the Development of Graft-Versus-Host Disease After Bone Marrow Transplantation. The New England Journal of Medicine. 1996;334:281–5. doi: 10.1056/NEJM199602013340501. [DOI (V体育安卓版)] [PubMed] [Google Scholar]
- 28.Goulmy E. Minor histocompatibility antigens: from transplantation problems to therapy of cancer. Hum Immunol. 2006;67:433–8. doi: 10.1016/j.humimm.2006.03.012. [DOI] [PubMed] [Google Scholar]
- 29.Murata M, Warren EH, Riddell SR. A human minor histocompatibility antigen resulting from differential expression due to a gene deletion. J Exp Med. 2003;197:1279–89. doi: 10.1084/jem.20030044. ["VSports app下载" DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Malarkannan S, Shih PP, Eden PA, Horng T, Zuberi AR, Christianson G, Roopenian D, Shastri N. The molecular and functional characterization of a dominant minor H antigen, H60. J Immunol. 1998;161:3501–9. [PubMed] [Google Scholar]
- 31.Bleakley M, Riddell SR. Molecules and mechanisms of the graft-versus-leukaemia effect. Nat Rev Cancer. 2004;4:371–80. doi: 10.1038/nrc1365. [DOI] [PubMed] [Google Scholar]
- 32.Kaplan DH, Anderson BE, McNiff JM, Jain D, Shlomchik MJ, Shlomchik WD. Target antigens determine graft-versus-host disease phenotype. J Immunol. 2004;173:5467–75. doi: 10.4049/jimmunol.173.9.5467. [DOI] [PubMed] [Google Scholar]
- 33.Riddell SR, Bleakley M, Nishida T, Berger C, Warren EH. Adoptive transfer of allogeneic antigen-specific T cells. Biol Blood Marrow Transplant. 2006;12:9–12. doi: 10.1016/j.bbmt.2005.10.025. ["VSports app下载" DOI] [PubMed] [Google Scholar]
- 34.Choi EY, Christianson GJ, Yoshimura Y, Jung N, Sproule TJ, Malarkannan S, Joyce S, Roopenian DC. Real-time T-cell profiling identifies H60 as a major minor histocompatibility antigen in murine graft-versus-host disease. Blood. 2002;100:4259–65. doi: 10.1182/blood-2002-05-1299. [DOI] [PubMed] [Google Scholar]
- 35.Choi EY, Christianson GJ, Yoshimura Y, Sproule TJ, Jung N, Joyce S, Roopenian DC. Immunodominance of H60 is caused by an abnormally high precursor T cell pool directed against its unique minor histocompatibility antigen peptide. Immunity. 2002;17:593–603. doi: 10.1016/s1074-7613(02)00428-4. [DOI (V体育官网入口)] [PubMed] [Google Scholar]
- 36.Fontaine P, Roy-Proulx G, Knafo L, Baron C, Roy DC, Perreault C. Adoptive transfer of minor histocompatibility antigen-specific T lymphocytes eradicates leukemia cells without causing graft-versus-host disease. Nat Med. 2001;7:789–94. doi: 10.1038/89907. [V体育安卓版 - DOI] [PubMed] [Google Scholar]
- 37.Dickinson AM, Wang XN, Sviland L, Vyth-Dreese FA, Jackson GH, Schumacher TN, Haanen JB, Mutis T, Goulmy E. In situ dissection of the graft-versus-host activities of cytotoxic T cells specific for minor histocompatibility antigens. Nat Med. 2002;8:410–4. doi: 10.1038/nm0402-410. [DOI] [PubMed] [Google Scholar]
- 38.de Bueger M, Bakker A, Bontkes H, van Rood JJ, Goulmy E. High frequencies of cytotoxic T cell precursors against minor histocompatibility antigens after HLA-identical BMT: absence of correlation with GVHD. Bone Marrow Transplantation. 1993:11. [PubMed] [Google Scholar]
- 39.Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, McKenna D, Le C, Defor TE, Burns LJ, Orchard PJ, Blazar BR, Wagner JE, Slungaard A, Weisdorf DJ, Okazaki IJ, McGlave PB. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–7. doi: 10.1182/blood-2004-07-2974. ["V体育官网" DOI] [PubMed] [Google Scholar]
- 40.Miller JS, Cooley S, Parham P, Farag SS, Verneris MR, McQueen KL, Guethlein LA, Trachtenberg EA, Haagenson M, Horowitz MM, Klein JP, Weisdorf DJ. Missing KIR-ligands is associated with less relapse and increased graft versus host disease (GVHD) following unrelated donor allogeneic HCT. Blood. 2007 doi: 10.1182/blood-2007-01-065383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Velardi A, Ruggeri L, Alessandro, Moretta, Moretta L. NK cells: a lesson from mismatched hematopoietic transplantation. Trends Immunol. 2002;23:438–44. doi: 10.1016/s1471-4906(02)02284-6. [DOI (VSports手机版)] [PubMed] [Google Scholar]
- 42.Petersdorf EW. Immunogenomics of unrelated hematopoietic cell transplantation. Curr Opin Immunol. 2006 doi: 10.1016/j.coi.2006.07.003. [DOI] [PubMed] [Google Scholar]
- 43.Hsu KC, Gooley T, Malkki M, Pinto-Agnello C, Dupont B, Bignon JD, Bornhauser M, Christiansen F, Gratwohl A, Morishima Y, Oudshoorn M, Ringden O, van Rood JJ, Petersdorf E. KIR Ligands and Prediction of Relapse after Unrelated Donor Hematopoietic Cell Transplantation for Hematologic Malignancy. Biol Blood Marrow Transplant. 2006;12:828–36. doi: 10.1016/j.bbmt.2006.04.008. ["V体育官网入口" DOI] [PubMed] [Google Scholar]
- 44.Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, Posati S, Rogaia D, Frassoni F, Aversa F, Martelli MF, Velardi A. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–100. doi: 10.1126/science.1068440. [VSports最新版本 - DOI] [PubMed] [Google Scholar]
- 45.Davies SM, Ruggieri L, DeFor T, Wagner JE, Weisdorf DJ, Miller JS, Velardi A, Blazar BR. Evaluation of KIR ligand incompatibility in mismatched unrelated donor hematopoietic transplants. Killer immunoglobulin-like receptor. Blood. 2002;100:3825–7. doi: 10.1182/blood-2002-04-1197. [DOI] [PubMed] [Google Scholar]
- 46.Hsu KC, Keever-Taylor CA, Wilton A, Pinto C, Heller G, Arkun K, O’Reilly RJ, Horowitz MM, Dupont B. Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes. Blood. 2005;105:4878–84. doi: 10.1182/blood-2004-12-4825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Antin JH, Ferrara JL. Cytokine dysregulation and acute graft-versus-host disease. Blood. 1992;80:2964–8. [PubMed] [Google Scholar]
- 48.Cavet J, Middleton PG, Segall M, Noreen H, Davies SM, Dickinson AM. Recipient tumor necrosis factor-alpha and interleukin-10 gene polymorphisms associate with early mortality and acute graft-versus-host disease severity in HLA-matched sibling bone marrow transplants. Blood. 1999;94:3941–6. [PubMed] [Google Scholar]
- 49.Dickinson AM, Charron D. Non-HLA immunogenetics in hematopoietic stem cell transplantation. Curr Opin Immunol. 2005;17:517–25. doi: 10.1016/j.coi.2005.07.017. [DOI] [PubMed] [Google Scholar]
- 50.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. N Engl J Med. 2003;349:2201–10. doi: 10.1056/NEJMoa022060. [DOI] [PubMed] [Google Scholar]
- 51.Mullighan CG, Bardy PG. New directions in the genomics of allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2007;13:127–44. doi: 10.1016/j.bbmt.2006.10.018. [DOI (V体育官网)] [PubMed] [Google Scholar]
- 52.Cavet J, Dickinson AM, Norden J, Taylor PR, Jackson GH, Middleton PG. Interferon-gamma and interleukin-6 gene polymorphisms associate with graft-versus-host disease in HLA-matched sibling bone marrow transplantation. Blood. 2001;98:1594–600. doi: 10.1182/blood.v98.5.1594. [DOI] [PubMed] [Google Scholar]
- 53.Brok HP, Heidt PJ, van der Meide PH, Zurcher C, Vossen JM. Interferon-gamma prevents graft-versus-host disease after allogeneic bone marrow transplantation in mice. J Immunol. 1993;151:6451–9. [PubMed (VSports注册入口)] [Google Scholar]
- 54.Yang YG, Dey BR, Sergio JJ, Pearson DA, Sykes M. Donor-derived interferon gamma is required for inhibition of acute graft-versus-host disease by interleukin 12. J Clin Invest. 1998;102:2126–35. doi: 10.1172/JCI4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Fishman D, Faulds G, Jeffery R, Mohamed-Ali V, Yudkin JS, Humphries S, Woo P. The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. J Clin Invest. 1998;102:1369–76. doi: 10.1172/JCI2629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.MacMillan ML, Radloff GA, Kiffmeyer WR, DeFor TE, Weisdorf DJ, Davies SM. High-producer interleukin-2 genotype increases risk for acute graft-versus-host disease after unrelated donor bone marrow transplantation. Transplantation. 2003;76:1758–62. doi: 10.1097/01.TP.0000095899.54052.89. [DOI] [PubMed] [Google Scholar]
- 57.Jordan WJ, Brookes PA, Szydlo RM, Goldman JM, Lechler RI, Ritter MA. IL-13 production by donor T cells is prognostic of acute graft-versus-host disease following unrelated donor stem cell transplantation. Blood. 2004;103:717–24. doi: 10.1182/blood-2003-01-0192. [DOI] [PubMed] [Google Scholar]
- 58.Holler E, Rogler G, Brenmoehl J, Hahn J, Herfarth H, Greinix H, Dickinson AM, Socie G, Wolff D, Fischer G, Jackson G, Rocha V, Steiner B, Eissner G, Marienhagen J, Schoelmerich J, Andreesen R. Prognostic significance of NOD2/CARD15 variants in HLA-identical sibling hematopoietic stem cell transplantation: effect on long-term outcome is confirmed in 2 independent cohorts and may be modulated by the type of gastrointestinal decontamination. Blood. 2006;107:4189–93. doi: 10.1182/blood-2005-09-3741. ["VSports在线直播" DOI] [PubMed] [Google Scholar]
- 59.Reddy P, Ferrara JL. Immunobiology of acute graft-versus-host disease. Blood Rev. 2003;17:187–94. doi: 10.1016/s0268-960x(03)00009-2. [DOI] [PubMed] [Google Scholar]
- 60.Gale RP, Bortin MM, van Bekkum DW, Biggs JC, Dicke KA, Gluckman E, Good RA, Hoffman RG, Kay HEM, Kersey JH, Marmont A, Masaoka T, Rimm AA, van Rood JJ, Zwaan FE. Risk factors for acute graft-versus-host disease. Br J Haematol. 1987;67:397–406. doi: 10.1111/j.1365-2141.1987.tb06160.x. [DOI] [PubMed] [Google Scholar]
- 61.Clift RA, Buckner CD, Appelbaum FR, Bearman SI, Petersen FB, Fisher LB, Anasetti C, Beatty P, Bensigner WI, Doney K, Hill RS, McDonald GB, Martin P, Sanders J, Singer J, Stewart P, Sullivan KM, Witherspoon R, Storb R, Hansen JA, Thomas ED. Allogeneic marrow transplantation in patients with acute myeloid leukemia in first remission: a randomized trial of two irradiation regimens. Blood. 1990;76:1867–71. [PubMed (V体育平台登录)] [Google Scholar]
- 62.Xun CQ, Thompson JS, Jennings CD, Brown SA, Widmer MB. 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;83:2360–7. [PubMed (V体育平台登录)] [Google Scholar]
- 63.Hill G, Ferrara J. The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 2000;95:2754–9. [PubMed] [Google Scholar]
- 64.Paris F, Fuks Z, Kang A, Capodieci P, Juan G, Ehleiter D, Haimovitz-Friedman A, Cordon-Cardo C, Kolesnick R. Endothelial apoptosis as the primary lesion initiating intestinal radiation damage in mice. Science. 2001;293:293–7. doi: 10.1126/science.1060191. [DOI] [PubMed] [Google Scholar]
- 65.Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JL. Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood. 1997;90:3204–13. [PubMed] [Google Scholar]
- 66.Matzinger P. The danger model: a renewed sense of self. Science. 2002;296:301–5. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
- 67.Shlomchik WD, Couzens MS, Tang CB, McNiff J, Robert ME, Liu J, Shlomchik MJ, Emerson SG. Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 1999;285:412–5. doi: 10.1126/science.285.5426.412. ["V体育ios版" DOI] [PubMed] [Google Scholar]
- 68.Murai M, Yoneyama H, Ezaki T, Suematsu M, Terashima Y, Harada A, Hamada H, Asakura H, Ishikawa H, Matsushima K. Peyer’s patch is the essential site in initiating murine acute and lethal graft-versus-host reaction. Nat Immunol. 2003;4:154–60. doi: 10.1038/ni879. [DOI] [PubMed] [Google Scholar]
- 69.Cooke K, Hill G, Crawford J, Bungard D, Brinson Y, Delmonte J, Jr, Ferrara J. Tumor necrosis factor-a production to lipopolysaccharide stimulation by donor cells predicts the severity of experimental acute graft versus host disease. J Clin Invest. 1998;102:1882–91. doi: 10.1172/JCI4285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Slavin S. New strategies for bone marrow transplantation. Curr Opin Immunol. 2000;12:542–51. doi: 10.1016/s0952-7915(00)00136-9. [DOI] [PubMed] [Google Scholar]
- 71.Teshima T, Ordemann R, Reddy P, Gagin S, Liu C, Cooke KR, Ferrara JL. Acute graft-versus-host disease does not require alloantigen expression on host epithelium. Nat Med. 2002;8:575–81. doi: 10.1038/nm0602-575. ["V体育2025版" DOI] [PubMed] [Google Scholar]
- 72.Jones SC, Murphy GF, Friedman TM, Korngold R. Importance of minor histocompatibility antigen expression by nonhematopoietic tissues in a CD4+ T cell-mediated graft-versus-host disease model. J Clin Invest. 2003;112:1880–6. doi: 10.1172/JCI19427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Reddy P, Maeda Y, Liu C, Krijanovski OI, Korngold R, Ferrara JL. A crucial role for antigen-presenting cells and alloantigen expression in graft-versus-leukemia responses. Nat Med. 2005;11:1244–9. doi: 10.1038/nm1309. [VSports最新版本 - DOI] [PubMed] [Google Scholar]
- 74.Arpinati M, Green CL, Heimfeld S, Heuser JE, Anasetti C. Granulocyte-colony stimulating factor mobilizes T helper 2-inducing dendritic cells. Blood. 2000;95:2484–90. [PubMed] [Google Scholar]
- 75.Auffermann-Gretzinger S, Lossos IS, Vayntrub TA, Leong W, Grumet FC, Blume KG, Stockerl-Goldstein KE, Levy R, Shizuru JA. Rapid establishment of dendritic cell chimerism in allogeneic hematopoietic cell transplant recipients. Blood. 2002;99:1442–8. doi: 10.1182/blood.v99.4.1442. [DOI (V体育2025版)] [PubMed] [Google Scholar]
- 76.MacDonald KP, Rowe V, Clouston AD, Welply JK, Kuns RD, Ferrara JL, Thomas R, Hill GR. Cytokine expanded myeloid precursors function as regulatory antigen-presenting cells and promote tolerance through IL-10-producing regulatory T cells. J Immunol. 2005;174:1841–50. doi: 10.4049/jimmunol.174.4.1841. [DOI] [PubMed] [Google Scholar]
- 77.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52. doi: 10.1038/32588. [DOI (VSports app下载)] [PubMed] [Google Scholar]
- 78.Duffner UA, Maeda Y, Cooke KR, Reddy P, Ordemann R, Liu C, Ferrara JL, Teshima T. Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease. J Immunol. 2004;172:7393–8. doi: 10.4049/jimmunol.172.12.7393. [DOI] [PubMed] [Google Scholar]
- 79.Sato K, Yamashita N, Baba M, Matsuyama T. Regulatory dendritic cells protect mice from murine acute graft-versus-host disease and leukemia relapse. Immunity. 2003;18:367–79. doi: 10.1016/s1074-7613(03)00055-4. [DOI] [PubMed] [Google Scholar]
- 80.Maeda Y, Reddy P, Lowler KP, Liu C, Bishop DK, Ferrara JL. Critical role of host gammadelta T cells in experimental acute graft-versus-host disease. Blood. 2005;106:749–55. doi: 10.1182/blood-2004-10-4087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Chorny A, Gonzalez-Rey E, Fernandez-Martin A, Ganea D, Delgado M. Vasoactive intestinal peptide induces regulatory dendritic cells that prevent acute graft-versus-host disease while maintaining the graft-versus-tumor response. Blood. 2006;107:3787–94. doi: 10.1182/blood-2005-11-4495. ["V体育官网入口" DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Macdonald KP, Kuns RD, Rowe V, Morris ES, Banovic T, Bofinger H, O’Sullivan B, Markey KA, Don AL, Thomas R, Hill GR. Effector and regulatory T cell function is differentially regulated by RelB within antigen presenting cells during GVHD. Blood. 2007 doi: 10.1182/blood-2007-01-067249. [DOI] [PubMed] [Google Scholar]
- 83.Paraiso KH, Ghansah T, Costello A, Engelman RW, Kerr WG. Induced SHIP Deficiency Expands Myeloid Regulatory Cells and Abrogates Graft-versus-Host Disease. J Immunol. 2007;178:2893–900. doi: 10.4049/jimmunol.178.5.2893. [DOI] [PubMed] [Google Scholar]
- 84.Merad M, Hoffmann P, Ranheim E, Slaymaker S, Manz MG, Lira SA, Charo I, Cook DN, Weissman IL, Strober S, Engleman EG. Depletion of host Langerhans cells before transplantation of donor alloreactive T cells prevents skin graft-versus-host disease. Nat Med. 2004;10:510–7. doi: 10.1038/nm1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Nachbaur D, Kircher B, Eisendle K, Latzer K, Haun M, Gastl G. Phenotype, function and chimaerism of monocyte-derived blood dendritic cells after allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2003;123:119–26. doi: 10.1046/j.1365-2141.2003.04588.x. [DOI] [PubMed] [Google Scholar]
- 86.Rowe V, Banovic T, MacDonald KP, Kuns R, Don AL, Morris ES, Burman AC, Bofinger HM, Clouston AD, Hill GR. Host B cells produce IL-10 following TBI and attenuate acute GVHD after allogeneic bone marrow transplantation. Blood. 2006;108:2485–92. doi: 10.1182/blood-2006-04-016063. [DOI] [PubMed] [Google Scholar]
- 87.Korngold R, Sprent J. Negative selection of T cells causing lethal graft-versus-host disease across minor histocompatibility barriers. Role of the H-2 complex. J Exp Med. 1980;151:1114–24. doi: 10.1084/jem.151.5.1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Beilhack A, Schulz S, Baker J, Beilhack GF, Wieland CB, Herman EI, Baker EM, Cao YA, Contag CH, Negrin RS. In vivo analyses of early events in acute graft-versus-host disease reveal sequential infiltration of T-cell subsets. Blood. 2005;106:1113–22. doi: 10.1182/blood-2005-02-0509. [DOI (VSports app下载)] [PMC free article] [PubMed] [Google Scholar]
- 89.Shlomchik WD. Antigen presentation in graft-vs-host disease. Exp Hematol. 2003;31:1187–97. doi: 10.1016/j.exphem.2003.09.017. [DOI (V体育2025版)] [PubMed] [Google Scholar]
- 90.Lechler R, Ng WF, Steinman RM. Dendritic cells in transplantation--friend or foe? Immunity. 2001;14:357–68. doi: 10.1016/s1074-7613(01)00116-9. [DOI] [PubMed] [Google Scholar]
- 91.Sayegh MH, Carpenter CB. Role of indirect allorecognition in allograft rejection. Int Rev Immunol. 1996;13:221–9. doi: 10.3109/08830189609061749. [DOI] [PubMed] [Google Scholar]
- 92.Anderson BE, McNiff JM, Jain D, Blazar BR, Shlomchik WD, Shlomchik MJ. Distinct roles for donor- and host-derived antigen-presenting cells and costimulatory molecules in murine chronic graft-versus-host disease: requirements depend on target organ. Blood. 2005;105:2227–34. doi: 10.1182/blood-2004-08-3032. [DOI] [PubMed] [Google Scholar]
- 93.Appleman LJ, Boussiotis VA. T cell anergy and costimulation. Immunol Rev. 2003;192:161–80. doi: 10.1034/j.1600-065x.2003.00009.x. [DOI] [PubMed] [Google Scholar]
- 94.Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol. 2002;2:116–26. doi: 10.1038/nri727. ["VSports注册入口" DOI] [PubMed] [Google Scholar]
- 95.Bromley SK, Iaboni A, Davis SJ, Whitty A, Green JM, Shaw AS, Weiss A, Dustin ML. The immunological synapse and CD28-CD80 interactions. Nat Immunol. 2001;2:1159–66. doi: 10.1038/ni737. [DOI] [PubMed] [Google Scholar]
- 96.Dustin ML. Role of adhesion molecules in activation signaling in T lymphocytes. J Clin Immunol. 2001;21:258–63. doi: 10.1023/a:1010927208180. [DOI] [PubMed] [Google Scholar]
- 97.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–48. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
- 98.Blazar BR, Taylor PA, Linsley PS, Vallera DA. In vivo blockade of CD28/CTLA4: B7/BB1 interaction with CTLA4-Ig reduces lethal murine graft-versus-host disease across the major histocompatibility complex barrier in mice. Blood. 1994;83:3815–25. ["VSports" PubMed] [Google Scholar]
- 99.Blazar BR, Taylor PA, Panoskaltsis-Mortari A, Buhlman J, Xu J, Flavell RA, Korngold R, Noelle R, Vallera DA. Blockade of CD40 ligand-CD40 interaction impairs CD4+ T cell-mediated alloreactivity by inhibiting mature donor T cell expansion and function after bone marrow transplantation. J Immunol. 1997;158:29–39. ["VSports在线直播" PubMed] [Google Scholar]
- 100.Blazar BR, Taylor PA, Panoskaltis-Mortari A, Gray GSDAV. Co-blockade of the LFA1:ICAM and CD28/CTLA4:B7 pathways is a highly effective means of preventing acute lethal graft-versus host disease induced by fully major histocompatibility complex-disparate donor grafts. Blood. 1995;85:2607–18. [VSports注册入口 - PubMed] [Google Scholar]
- 101.Blazar BR, Kwon BS, Panoskaltsis-Mortari A, Kwak KB, Peschon JJ, Taylor PA. Ligation of 4-1BB (CDw137) regulates graft-versus-host disease, graft-versus-leukemia, and graft rejection in allogeneic bone marrow transplant recipients. J Immunol. 2001;166:3174–83. doi: 10.4049/jimmunol.166.5.3174. [DOI] [PubMed] [Google Scholar]
- 102.Blazar BR, Sharpe AH, Chen AI, Panoskaltsis-Mortari A, Lees C, Akiba H, Yagita H, Killeen N, Taylor PA. Ligation of OX40 (CD134) regulates graft-versus-host disease (GVHD) and graft rejection in allogeneic bone marrow transplant recipients. Blood. 2003;101:3741–8. doi: 10.1182/blood-2002-10-3048. [DOI] [PubMed] [Google Scholar]
- 103.Blazar BR, Carreno BM, Panoskaltsis-Mortari A, Carter L, Iwai Y, Yagita H, Nishimura H, Taylor PA. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J Immunol. 2003;171:1272–7. doi: 10.4049/jimmunol.171.3.1272. ["V体育官网入口" DOI] [PubMed] [Google Scholar]
- 104.Csencsits KL, Bishop DK. Contrasting alloreactive CD4+ and CD8+ T cells: there’s more to it than MHC restriction. Am J Transplant. 2003;3:107–15. doi: 10.1034/j.1600-6143.2003.00036.x. ["VSports app下载" DOI] [PubMed] [Google Scholar]
- 105.Korngold R, Sprent J. Features of T cells causing H-2-restricted lethal graft-vs.-host disease across minor histocompatibility barriers. J Exp Med. 1982;155:872–83. doi: 10.1084/jem.155.3.872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Korngold R, Sprent J. Surface markers of T cells causing lethal graft-vs-host disease to class I vs class II H-2 differences. Journal of Immunology. 1985;135:3004–10. [PubMed] [Google Scholar]
- 107.Korngold R, Sprent J. T cell subsets and graft-versus-host disease. Transplantation. 1987;44:335–9. doi: 10.1097/00007890-198709000-00002. [DOI] [PubMed] [Google Scholar]
- 108.Wu CJ, Ritz J. Induction of tumor immunity following allogeneic stem cell transplantation. Adv Immunol. 2006;90:133–73. doi: 10.1016/S0065-2776(06)90004-2. [DOI] [PubMed] [Google Scholar]
- 109.Spierings E, Drabbels J, Hendriks M, Pool J, Spruyt-Gerritse M, Claas F, Goulmy E. A uniform genomic minor histocompatibility antigen typing methodology and database designed to facilitate clinical applications. PLoS ONE. 2006;1:e42. doi: 10.1371/journal.pone.0000042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wekerle T, Kurtz J, Ito H, Ronquillo JV, Dong V, Zhao G, Shaffer J, Sayegh MH, Sykes M. Allogeneic bone marrow transplantation with co-stimulatory blockade induces macrochimerism and tolerance without cytoreductive host treatment. Nat Med. 2000;6:464–9. doi: 10.1038/74731. [DOI (VSports最新版本)] [PubMed] [Google Scholar]
- 111.Wekerle T, Sayegh MH, Hill J, Zhao Y, Chandraker A, Swenson KG, Zhao G, Sykes M. Extrathymic T cell deletion and allogeneic stem cell engraftment induced with costimulatory blockade is followed by central T cell tolerance. J Exp Med. 1998;187:2037–44. doi: 10.1084/jem.187.12.2037. [DOI (VSports最新版本)] [PMC free article] [PubMed] [Google Scholar]
- 112.Sykes M. Mixed chimerism and transplant tolerance. Immunity. 2001;14:417–24. doi: 10.1016/s1074-7613(01)00122-4. [DOI] [PubMed] [Google Scholar]
- 113.Wells AD, Li XC, Li Y, Walsh MC, Zheng XX, Wu Z, Nunez G, Tang A, Sayegh M, Hancock WW, Strom TB, Turka LA. Requirement for T-cell apoptosis in the induction of peripheral transplantation tolerance. Nat Med. 1999;5:1303–7. doi: 10.1038/15260. [DOI] [PubMed] [Google Scholar]
- 114.Wells AD, Li XC, Strom TB, Turka LA. The role of peripheral T-cell deletion in transplantation tolerance. Philos Trans R Soc Lond B Biol Sci. 2001;356:617–23. doi: 10.1098/rstb.2001.0845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Lechler RI, Garden OA, Turka LA. The complementary roles of deletion and regulation in transplantation tolerance. Nat Rev Immunol. 2003;3:147–58. doi: 10.1038/nri1002. [DOI] [PubMed] [Google Scholar]
- 116.Zheng L, Fisher G, Miller R, Peschon J, Lynch D, Lenardo M. Induction of apoptosis in mature T cells by tumor necrosis factor. Nature. 1995;377:348–51. doi: 10.1038/377348a0. [DOI] [PubMed] [Google Scholar]
- 117.Kishimoto K, Sandner S, Imitola J, Sho M, Li Y, Langmuir PB, Rothstein DM, Strom TB, Turka LA, Sayegh MH. Th1 cytokines, programmed cell death, and alloreactive T cell clone size in transplant tolerance. J Clin Invest. 2002;109:1471–9. doi: 10.1172/JCI14947. [VSports app下载 - DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Min CK, Maeda Y, Lowler K, Liu C, Clouthier S, Lofthus D, Weisiger E, Ferrara JL, Reddy P. Paradoxical effects of interleukin-18 on the severity of acute graft-versus-host disease mediated by CD4+ and CD8+ T-cell subsets after experimental allogeneic bone marrow transplantation. Blood. 2004;104:3393–9. doi: 10.1182/blood-2004-02-0763. ["V体育安卓版" DOI] [PubMed] [Google Scholar]
- 119.Zhang X, Brunner T, Carter L, Dutton RW, Rogers P. Unequal Death in T Helper Cell (Th)1 and Th2 Effectors: Th1, but not Th2, Effectors Undergo Rapid Fas/FasL-mediated Apoptosis. Journal of Experimental Medicine. 1997;185:1837–49. doi: 10.1084/jem.185.10.1837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Siegel RM, Chan FK, Chun HJ, Lenardo MJ. The multifaceted role of Fas signaling in immune cell homeostasis and autoimmunity. Nat Immunol. 2000;1:469–74. doi: 10.1038/82712. [DOI] [PubMed] [Google Scholar]
- 121.Combadiere B, Reis e Sousa C, Trageser C, Zheng LX, Kim CR, Lenardo MJ. Differential TCR signaling regulates apoptosis and immunopathology during antigen responses in vivo. Immunity. 1998;9:305–13. doi: 10.1016/s1074-7613(00)80613-5. [DOI] [PubMed] [Google Scholar]
- 122.Drobyski WR, Komorowski R, Logan B, Gendelman M. Role of the passive apoptotic pathway in graft-versus-host disease. J Immunol. 2002;169:1626–33. doi: 10.4049/jimmunol.169.3.1626. [V体育2025版 - DOI] [PubMed] [Google Scholar]
- 123.Bonini C, Verzeletti S, Servida P, Rossini S, Traversari C, Ferrari G, Nobili N, Mavilio F, Bordignon C. Transfer of the HSV-tk gene into donor peripheral blood lymphocytes for in vivo immunomodulation of donor anti-tumor immunity after allo-BMT. Blood. 1994;83:1988. [Google Scholar]
- 124.Bonini C, Ferrari G, Verzeletti S, Servida P, Zappone E. HSV-TK Gene Transfer into Donor Lymphocytes for Control of Allogeneic Graft-Versus-Leukemia. Science. 1997;276:1719–24. doi: 10.1126/science.276.5319.1719. [DOI] [PubMed] [Google Scholar]
- 125.Drobyski WR, Gendelman M. Regulation of alloresponses after bone marrow transplantation using donor T cells expressing a thymidine kinase suicide gene. Leuk Lymphoma. 2002;43:2011–6. doi: 10.1080/1042819021000016041. [DOI] [PubMed] [Google Scholar]
- 126.Bondanza A, Valtolina V, Magnani Z, Ponzoni M, Fleischhauer K, Bonyhadi M, Traversari C, Sanvito F, Toma S, Radrizzani M, La Seta-Catamancio S, Ciceri F, Bordignon C, Bonini C. Suicide gene therapy of graft-versus-host disease induced by central memory human T lymphocytes. Blood. 2006;107:1828–36. doi: 10.1182/blood-2005-09-3716. ["V体育平台登录" DOI] [PubMed] [Google Scholar]
- 127.Traversari C, Marktel S, Magnani Z, Mangia P, Russo V, Ciceri F, Bonini C, Bordignon C. The potential immunogenicity of the TK suicide gene does not prevent full clinical benefit associated with the use of TK-transduced donor lymphocytes in HSCT for hematologic malignancies. Blood. 2007 doi: 10.1182/blood-2006-04-015230. [DOI (V体育2025版)] [PubMed] [Google Scholar]
- 128.Chen BJ, Cui X, Liu C, Chao NJ. Prevention of graft-versus-host disease while preserving graft-versus-leukemia effect after selective depletion of host-reactive T cells by photodynamic cell purging process. Blood. 2002;99:3083–8. doi: 10.1182/blood.v99.9.3083. [DOI] [PubMed] [Google Scholar]
- 129.Chen BJ, Chen Y, Cui X, Fidler JM, Chao NJ. Mechanisms of tolerance induced by PG490-88 in a bone marrow transplantation model. Transplantation. 2002;73:115–21. doi: 10.1097/00007890-200201150-00022. [DOI] [PubMed] [Google Scholar]
- 130.Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, Shlomchik WD. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest. 2003;112:101–8. doi: 10.1172/JCI17601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Chen BJ, Cui X, Sempowski GD, Liu C, Chao NJ. Transfer of allogeneic CD62L- memory T cells without graft-versus-host disease. Blood. 2004;103:1534–41. doi: 10.1182/blood-2003-08-2987. [DOI] [PubMed] [Google Scholar]
- 132.Zhang Y, Joe G, Zhu J, Carroll R, Levine B, Hexner E, June C, Emerson SG. Dendritic cell-activated CD44hiCD8+ T cells are defective in mediating acute graft-versus-host disease but retain graft-versus-leukemia activity. Blood. 2004;103:3970–8. doi: 10.1182/blood-2003-09-3135. [DOI] [PubMed] [Google Scholar]
- 133.Maeda Y, Tawara I, Teshima T, Liu C, Hashimoto D, Matsuoka K, Tanimoto M, Reddy P. Lymphopenia-induced proliferation of donor T cells reduces their capacity for causing acute graft-versus-host disease. Exp Hematol. 2007;35:274–86. doi: 10.1016/j.exphem.2006.10.010. [DOI] [PubMed] [Google Scholar]
- 134.Taylor PA, Panoskaltsis-Mortari A, Swedin JM, Lucas PJ, Gress RE, Levine BL, June CH, Serody JS, Blazar BR. L-Selectin(hi) but not the L-selectin(lo) CD4+25+ T-regulatory cells are potent inhibitors of GVHD and BM graft rejection. Blood. 2004;104:3804–12. doi: 10.1182/blood-2004-05-1850. [DOI] [PubMed] [Google Scholar]
- 135.Ermann J, Hoffmann P, Edinger M, Dutt S, Blankenberg FG, Higgins JP, Negrin RS, Fathman CG, Strober S. Only the CD62L+ subpopulation of CD4+CD25+ regulatory T cells protects from lethal acute GVHD. Blood. 2005;105:2220–6. doi: 10.1182/blood-2004-05-2044. [DOI] [PubMed] [Google Scholar]
- 136.Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Alloreactive memory T cells are responsible for the persistence of graft-versus-host disease. J Immunol. 2005;174:3051–8. doi: 10.4049/jimmunol.174.5.3051. [DOI] [PubMed] [Google Scholar]
- 137.Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat Med. 2005;11:1299–305. doi: 10.1038/nm1326. [DOI] [PubMed] [Google Scholar]
- 138.Wu Z, Bensinger SJ, Zhang J, Chen C, Yuan X, Huang X, Markmann JF, Kassaee A, Rosengard BR, Hancock WW, Sayegh MH, Turka LA. Homeostatic proliferation is a barrier to transplantation tolerance. Nat Med. 2004;10:87–92. doi: 10.1038/nm965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Blazar BR, Taylor PA. Regulatory T cells. Biol Blood Marrow Transplant. 2005;11:46–9. doi: 10.1016/j.bbmt.2004.11.005. [DOI] [PubMed] [Google Scholar]
- 140.Cohen JL, Boyer O. The role of CD4(+)CD25(hi) regulatory T cells in the physiopathogeny of graft-versus-host disease. Curr Opin Immunol. 2006 doi: 10.1016/j.coi.2006.07.007. [DOI] [PubMed] [Google Scholar]
- 141.Johnson BD, Konkol MC, Truitt RL. CD25+ immunoregulatory T-cells of donor origin suppress alloreactivity after BMT. Biol Blood Marrow Transplant. 2002;8:525–35. doi: 10.1053/bbmt.2002.v8.pm12434947. [DOI] [PubMed] [Google Scholar]
- 142.Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med. 2002;196:389–99. doi: 10.1084/jem.20020399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Cohen JL, Trenado A, Vasey D, Klatzmann D, Salomon BL. CD4(+)CD25(+) immunoregulatory T Cells: new therapeutics for graft-versus-host disease. J Exp Med. 2002;196:401–6. doi: 10.1084/jem.20020090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Roncarolo MG. The Role of Interleukin-10 in Transplantation and GVHD. In: Ferrara JLM, Deeg HJ, Burakoff SJ, editors. Graft-vs-Host Disease. New York: Marcel Dekker, Inc; 1997. pp. 693–715. [VSports - Google Scholar]
- 145.Young KJ, DuTemple B, Phillips MJ, Zhang L. Inhibition of graft-versus-host disease by double-negative regulatory T cells. J Immunol. 2003;171:134–41. doi: 10.4049/jimmunol.171.1.134. [DOI] [PubMed] [Google Scholar]
- 146.Zeng D, Lewis D, Dejbakhsh-Jones S, Lan F, Garcia-Ojeda M, Sibley R, Strober S. Bone marrow NK1.1(−) and NK1.1(+) T cells reciprocally regulate acute graft versus host disease. J Exp Med. 1999;189:1073–81. doi: 10.1084/jem.189.7.1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Lowsky R, Takahashi T, Liu YP, Dejbakhsh-Jones S, Grumet FC, Shizuru JA, Laport GG, Stockerl-Goldstein KE, Johnston LJ, Hoppe RT, Bloch DA, Blume KG, Negrin RS, Strober S. Protective conditioning for acute graft-versus-host disease. N Engl J Med. 2005;353:1321–31. doi: 10.1056/NEJMoa050642. [DOI] [PubMed] [Google Scholar]
- 148.Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S, Negrin RS. CD4(+)CD25(+) regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9:1144–50. doi: 10.1038/nm915. [DOI] [PubMed] [Google Scholar]
- 149.Nguyen VH, Zeiser R, Dasilva DL, Chang DS, Beilhack A, Contag CH, Negrin RS. In vivo dynamics of regulatory T-cell trafficking and survival predict effective strategies to control graft-versus-host disease following allogeneic transplantation. Blood. 2007;109:2649–56. doi: 10.1182/blood-2006-08-044529. [DOI] [PubMed] [Google Scholar]
- 150.Stanzani M, Martins SL, Saliba RM, St John LS, Bryan S, Couriel D, McMannis J, Champlin RE, Molldrem JJ, Komanduri KV. CD25 expression on donor CD4+ or CD8+ T cells is associated with an increased risk of graft-versus-host disease following HLA-identical stem cell transplantation in humans. Blood. 2003 doi: 10.1182/blood-2003-06-2085. [V体育ios版 - DOI] [PubMed] [Google Scholar]
- 151.Miura Y, Thoburn CJ, Bright EC, Phelps ML, Shin T, Matsui EC, Matsui WH, Arai S, Fuchs EJ, Vogelsang GB, Jones RJ, Hess AD. Association of Foxp3 regulatory gene expression with graft-versus-host disease. Blood. 2004;104:2187–93. doi: 10.1182/blood-2004-03-1040. [VSports最新版本 - DOI] [PubMed] [Google Scholar]
- 152.Zorn E, Kim HT, Lee SJ, Floyd BH, Litsa D, Arumugarajah S, Bellucci R, Alyea EP, Antin JH, Soiffer RJ, Ritz J. Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease. Blood. 2005;106:2903–11. doi: 10.1182/blood-2005-03-1257. ["VSports在线直播" DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ziegler SF. FOXP3: of mice and men. Annu Rev Immunol. 2006;24:209–26. doi: 10.1146/annurev.immunol.24.021605.090547. [DOI] [PubMed] [Google Scholar]
- 154.Hashimoto D, Asakura S, Miyake S, Yamamura T, Van Kaer L, Tanimoto M, Teshima T. Host NKT cells promote Th2 polarization of donor T cells and regulate acute GVHD after experimental BMT via a STAT6-dependent mechanism. Blood. 2003;102:191a. [Google Scholar]
- 155.Morris ES, MacDonald KP, Rowe V, Banovic T, Kuns RD, Don AL, Bofinger HM, Burman AC, Olver SD, Kienzle N, Porcelli SA, Pellicci DG, Godfrey DI, Smyth MJ, Hill GR. NKT cell-dependent leukemia eradication following stem cell mobilization with potent G-CSF analogs. J Clin Invest. 2005;115:3093–103. doi: 10.1172/JCI25249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Morris ES, MacDonald KP, Hill GR. Stem cell mobilization with G-CSF analogs: a rational approach to separate GVHD and GVL? Blood. 2006;107:3430–5. doi: 10.1182/blood-2005-10-4299. [DOI] [PubMed] [Google Scholar]
- 157.Wang JW, Howson JM, Ghansah T, Desponts C, Ninos JM, May SL, Nguyen KH, Toyama-Sorimachi N, Kerr WG. Influence of SHIP on the NK repertoire and allogeneic bone marrow transplantation. Science. 2002;295:2094–7. doi: 10.1126/science.1068438. ["VSports app下载" DOI] [PubMed] [Google Scholar]
- 158.Ferrara J, Krenger W. Graft Versus Host Disease: The Influence of Type 1 and Type 2 T Cell Cytokines. Transfusion Medicine Reviews. 1998;12:1–17. doi: 10.1016/s0887-7963(98)80085-0. [DOI (VSports最新版本)] [PubMed] [Google Scholar]
- 159.Ferrara JLM. The cytokine storm of acute graft-versus host disease. Haematol Rev. 1994;8:27. ["V体育官网" Google Scholar]
- 160.Reddy P. Pathophysiology of acute graft-versus-host disease. Hematol Oncol. 2003;21:149–61. doi: 10.1002/hon.716. [DOI] [PubMed] [Google Scholar]
- 161.Ratanatharathorn V, Nash RA, Przepiorka D, et al. Phase III study comparing methotrexate and tacrolimus (prpgraf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation. Blood. 1998;92:2303–14. [PubMed] [Google Scholar]
- 162.Liu EH, Siegel RM, Harlan DM, O’Shea JJ. T cell-directed therapies: lessons learned and future prospects. Nat Immunol. 2007;8:25–30. doi: 10.1038/ni1429. [VSports手机版 - DOI] [PubMed] [Google Scholar]
- 163.Zeiser R, Nguyen VH, Beilhack A, Buess M, Schulz S, Baker J, Contag CH, Negrin RS. Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood. 2006;108:390–9. doi: 10.1182/blood-2006-01-0329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Zhang H, Chua KS, Guimond M, Kapoor V, Brown MV, Fleisher TA, Long LM, Bernstein D, Hill BJ, Douek DC, Berzofsky JA, Carter CS, Read EJ, Helman LJ, Mackall CL. Lymphopenia and interleukin-2 therapy alter homeostasis of CD4+CD25+ regulatory T cells. Nat Med. 2005;11:1238–43. doi: 10.1038/nm1312. [DOI] [PubMed] [Google Scholar]
- 165.Liston A, Rudensky AY. Thymic development and peripheral homeostasis of regulatory T cells. Curr Opin Immunol. 2007;19:176–85. doi: 10.1016/j.coi.2007.02.005. [DOI] [PubMed] [Google Scholar]
- 166.Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, Rudensky AY. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445:771–5. doi: 10.1038/nature05543. [DOI] [PubMed] [Google Scholar]
- 167.Brok HP, Vossen JM, Heidt PJ. Interferon-gamma-mediated prevention of graft-versus-host disease: development of immune competent and allo-tolerant T cells in chimeric mice. Bone Marrow Transplant. 1997;19:601–6. doi: 10.1038/sj.bmt.1700707. [DOI] [PubMed] [Google Scholar]
- 168.Ellison CA, Fischer JM, HayGlass KT, Gartner JG. Murine graft-versus-host disease in an F1-hybrid model using IFN-gamma gene knockout donors. J Immunol. 1998;161:631–40. [PubMed] [Google Scholar]
- 169.Burman AC, Banovic TD, Kuns RD, Clouston AD, Stanley AC, Morris ES, Rowe V, Bofinger H, Skoczylas R, Raffelt N, Fahy O, McColl SR, Engwerda CR, Macdonald KP, Hill GR. IFN{gamma} differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract. Blood. 2007 doi: 10.1182/blood-2006-12-063982. [DOI] [PubMed] [Google Scholar]
- 170.Sykes M, Romick ML, Hoyles KA, Sachs DH. In vivo administration of interleukin 2 plus T cell-depleted syngeneic marrow prevents graft-versus-host disease mortality and permits alloengraftment. J Exp Med. 1990;171:645–58. doi: 10.1084/jem.171.3.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Sykes M, Romick ML, Sachs DH. Interleukin 2 prevents graft-versus-host disease while preserving the graft-versus-leukemia effect of allogeneic T cells. Proc Natl Acad Sci USA. 1990;87:5633–7. doi: 10.1073/pnas.87.15.5633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Yang Y-G, Sergio JJ, Pearson DA, Szot GL, Shimizu A, Sykes M. Interleukin-12 preserves the graft-versus-leukemia effect of allogeneic CD8 T cells while inhibiting CD4-dependent graft-versus-host disease in mice. Blood. 1997;90:4651–60. [PubMed] [Google Scholar]
- 173.Sykes M, Pearson DA, Taylor PA, Szot GL, Goldman SJ, Blazar BR. Dose and timing of interleukin (IL)-12 and timing and type of total-body irradiation: effects on graft-vs.-host disease inhibition and toxicity of exogenous IL-12 in murine bone marrow transplant recipients. Biol Blood Marrow Transplant. 1999;5:277–84. doi: 10.1016/s1083-8791(99)70002-9. [DOI] [PubMed] [Google Scholar]
- 174.Reddy P. Interleukin-18: recent advances. Curr Opin Hematol. 2004;11:405–10. doi: 10.1097/01.moh.0000141926.95319.42. [DOI] [PubMed] [Google Scholar]
- 175.Sykes M, Harty MW, Szot GL, Pearson DA. Interleukin-2 inhibits graft-versus-host disease-promoting activity of CD4+ cells while preserving CD4- and CD8-mediated graft-versus-leukemia effects. Blood. 1994;83:2560–9. ["V体育平台登录" PubMed] [Google Scholar]
- 176.Sykes M, Szot GL, Nguyen PL, Pearson DA. Interleukin-12 inhibits murine graft-versus-host disease. Blood. 1995;86:2429–38. [PubMed] [Google Scholar]
- 177.Reddy P, Teshima T, Kukuruga M, Ordemann R, Liu C, Lowler K, Ferrara JL. Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis. J Exp Med. 2001;194:1433–40. doi: 10.1084/jem.194.10.1433. ["VSports最新版本" DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Fowler DH, Kurasawa K, Smith R, Eckhaus MA, Gress RE. Donor CD4-enriched cells of Th2 cytokine phenotype regulate graft-versus-host disease without impairing allogeneic engraftment in sublethally irradiated mice. Blood. 1994;84:3540. [PubMed] [Google Scholar]
- 179.Krenger W, Snyder KM, Byon CH, Falzarano G, Ferrara JLM. Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease. J Immunol. 1995;155:585–93. [PubMed] [Google Scholar]
- 180.Pan L, Delmonte J, Jalonen C, Ferrara J. Pretreatment of donor mice with granulocyte colony-stimulating factor polarizes donor T-lymphocytes toward type-2 cytokine production and reduces severity of experimental graft-versus-host disease. Blood. 1995;86:4422–9. [VSports app下载 - PubMed] [Google Scholar]
- 181.Hill GR, Cooke KR, Teshima T, Crawford JM, Keith JC, Jr, Brinson YS, Bungard D, Ferrara JL. Interleukin-11 promotes T cell polarization and prevents acute graft-versus-host disease after allogeneic bone marrow transplantation. J Clin Invest. 1998;102:115–23. doi: 10.1172/JCI3132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Reddy P, Teshima T, Hildebrandt G, Williams DL, Liu C, Cooke KR, Ferrara JL. Pretreatment of donors with interleukin-18 attenuates acute graft-versus-host disease via STAT6 and preserves graft-versus-leukemia effects. Blood. 2003;101:2877–85. doi: 10.1182/blood-2002-08-2566. [DOI (VSports在线直播)] [PubMed] [Google Scholar]
- 183.Foley JE, Jung U, Miera A, Borenstein T, Mariotti J, Eckhaus M, Bierer BE, Fowler DH. Ex vivo rapamycin generates donor Th2 cells that potently inhibit graft-versus-host disease and graft-versus-tumor effects via an IL-4-dependent mechanism. J Immunol. 2005;175:5732–43. doi: 10.4049/jimmunol.175.9.5732. [DOI] [PubMed] [Google Scholar]
- 184.Jung U, Foley JE, Erdmann AA, Toda Y, Borenstein T, Mariotti J, Fowler DH. Ex Vivo Rapamycin Generates Th1/Tc1 or Th2/Tc2 Effector T Cells With Enhanced In Vivo Function and Differential Sensitivity to Post-transplant Rapamycin Therapy. Biol Blood Marrow Transplant. 2006;12:905–18. doi: 10.1016/j.bbmt.2006.05.014. [V体育官网 - DOI] [PubMed] [Google Scholar]
- 185.Fowler DH, Gress RE. Th2 and Tc2 cells in the regulation of GVHD, GVL, and graft rejection: considerations for the allogeneic transplantation therapy of leukemia and lymphoma. Leuk Lymphoma. 2000;38:221–34. doi: 10.3109/10428190009087014. [DOI] [PubMed] [Google Scholar]
- 186.Nikolic B, Lee S, Bronson R, Grusby M, Sykes M. Th1 and Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets. J Clin Invest. 2000;105:1289–98. doi: 10.1172/JCI7894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Blazar BR, Taylor PA, Smith S, Vallera DA. Interleukin-10 administration decreases survival in murine recipients of major histocompatibility complex disparate donor bone marrow grafts. Blood. 1995;85:842–51. [PubMed] [Google Scholar]
- 188.Banovic T, MacDonald KP, Morris ES, Rowe V, Kuns R, Don A, Kelly J, Ledbetter S, Clouston AD, Hill GR. TGF-beta in allogeneic stem cell transplantation: friend or foe? Blood. 2005;106:2206–14. doi: 10.1182/blood-2005-01-0062. [DOI] [PubMed] [Google Scholar]
- 189.Alpdogan O, Schmaltz C, Muriglan SJ, Kappel BJ, Perales MA, Rotolo JA, Halm JA, Rich BE, van den Brink MR. Administration of interleukin-7 after allogeneic bone marrow transplantation improves immune reconstitution without aggravating graft-versus-host disease. Blood. 2001;98:2256–65. doi: 10.1182/blood.v98.7.2256. [DOI] [PubMed] [Google Scholar]
- 190.Alpdogan O, Muriglan SJ, Eng JM, Willis LM, Greenberg AS, Kappel BJ, van den Brink MR. IL-7 enhances peripheral T cell reconstitution after allogeneic hematopoietic stem cell transplantation. J Clin Invest. 2003;112:1095–107. doi: 10.1172/JCI17865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Sinha ML, Fry TJ, Fowler DH, Miller G, Mackall CL. Interleukin 7 worsens graft-versus-host disease. Blood. 2002;100:2642–9. doi: 10.1182/blood-2002-04-1082. ["V体育平台登录" DOI] [PubMed] [Google Scholar]
- 192.Gendelman M, Hecht T, Logan B, Vodanovic-Jankovic S, Komorowski R, Drobyski WR. Host conditioning is a primary determinant in modulating the effect of IL-7 on murine graft-versus-host disease. J Immunol. 2004;172:3328–36. doi: 10.4049/jimmunol.172.5.3328. [DOI] [PubMed] [Google Scholar]
- 193.Weaver CT, Murphy KM. T-cell subsets: the more the merrier. Curr Biol. 2007;17:R61–3. doi: 10.1016/j.cub.2006.12.015. [DOI] [PubMed] [Google Scholar]
- 194.Wysocki CA, Panoskaltsis-Mortari A, Blazar BR, Serody JS. Leukocyte migration and graft-versus-host disease. Blood. 2005;105:4191–9. doi: 10.1182/blood-2004-12-4726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Cyster JG. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol. 2005;23:127–59. doi: 10.1146/annurev.immunol.23.021704.115628. [DOI] [PubMed] [Google Scholar]
- 196.Serody JS, Burkett SE, Panoskaltsis-Mortari A, Ng-Cashin J, McMahon E, Matsushima GK, Lira SA, Cook DN, Blazar BR. T-lymphocyte production of macrophage inflammatory protein-1alpha is critical to the recruitment of CD8(+) T cells to the liver, lung, and spleen during graft-versus-host disease. Blood. 2000;96:2973–80. [PubMed] [Google Scholar]
- 197.Mapara MY, Leng C, Kim YM, Bronson R, Lokshin A, Luster A, Sykes M. Expression of chemokines in GVHD target organs is influenced by conditioning and genetic factors and amplified by GVHR. Biol Blood Marrow Transplant. 2006;12:623–34. doi: 10.1016/j.bbmt.2006.02.005. [DOI] [PubMed] [Google Scholar]
- 198.Duffner U, Lu B, Hildebrandt GC, Teshima T, Williams DL, Reddy P, Ordemann R, Clouthier SG, Lowler K, Liu C, Gerard C, Cooke KR, Ferrara JL. Role of CXCR3-induced donor T-cell migration in acute GVHD. Exp Hematol. 2003;31:897–902. doi: 10.1016/s0301-472x(03)00198-x. [V体育官网入口 - DOI] [PubMed] [Google Scholar]
- 199.Wysocki CA, Burkett SB, Panoskaltsis-Mortari A, Kirby SL, Luster AD, McKinnon K, Blazar BR, Serody JS. Differential roles for CCR5 expression on donor T cells during graft-versus-host disease based on pretransplant conditioning. J Immunol. 2004;173:845–54. doi: 10.4049/jimmunol.173.2.845. [DOI (VSports)] [PubMed] [Google Scholar]
- 200.Murai M, Yoneyama H, Harada A, Yi Z, Vestergaard C, Guo B, Suzuki K, Asakura H, Matsushima K. Active participation of CCR5(+)CD8(+) T lymphocytes in the pathogenesis of liver injury in graft-versus-host disease. J Clin Invest. 1999;104:49–57. doi: 10.1172/JCI6642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Wysocki C, Burkett S, Chwastiak K, Taylor P, Blazar BR, Serody JS. CCR5 expression on CD4+CD25+ regulatory T cells plays a critical role in their ability to prevent GVHD. Blood. 2003;102:#517. ["V体育安卓版" Google Scholar]
- 202.Waldman E, Lu SX, Hubbard VM, Kochman AA, Eng JM, Terwey TH, Muriglan SJ, Kim TD, Heller G, Murphy GF, Liu C, Alpdogan O, van den Brink MR. Absence of beta7 integrin results in less graft-versus-host disease because of decreased homing of alloreactive T cells to intestine. Blood. 2006;107:1703–11. doi: 10.1182/blood-2005-08-3445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Welniak LA, Kuprash DV, Tumanov AV, Panoskaltsis-Mortari A, Blazar BR, Sun K, Nedospasov SA, Murphy WJ. Peyer’s patches are not required for acute graft-versus-host disease after myeloablative conditioning and murine allogeneic bone marrow transplantation. Blood. 2005 doi: 10.1182/blood-2004-11-4565. [VSports手机版 - DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Kim YM, Sachs T, Asavaroengchai W, Bronson R, Sykes M. Graft-versus-host disease can be separated from graft-versus-lymphoma effects by control of lymphocyte trafficking with FTY720. J Clin Invest. 2003;111:659–69. doi: 10.1172/JCI16950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Lee RS, Kuhr CS, Sale GE, Zellmer E, Hogan WJ, Storb R, Little MT. FTY720 does not abrogate acute graft-versus-host disease in the dog leukocyte antigen-nonidentical unrelated canine model. Transplantation. 2003;76:1155–8. doi: 10.1097/01.TP.0000083891.14089.B8. ["VSports在线直播" DOI] [PubMed] [Google Scholar]
- 206.Sallusto F, Mackay CR, Lanzavecchia A. The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol. 2000;18:593–620. doi: 10.1146/annurev.immunol.18.1.593. [DOI (VSports app下载)] [PubMed] [Google Scholar]
- 207.Sackstein R. A revision of Billingham’s tenets: the central role of lymphocyte migration in acute graft-versus-host disease. Biol Blood Marrow Transplant. 2006;12:2–8. doi: 10.1016/j.bbmt.2005.09.015. [V体育平台登录 - DOI] [PubMed] [Google Scholar]
- 208.Kagi D, Vignaux F, Ledermann B, Burki K, Depraetere V, Nagata S, Hengartner H, Golstein P. Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity. Science. 1994;265:528–30. doi: 10.1126/science.7518614. ["V体育安卓版" DOI] [PubMed] [Google Scholar]
- 209.van den Brink MR, Burakoff SJ. Cytolytic pathways in haematopoietic stem-cell transplantation. Nat Rev Immunol. 2002;2:273–81. doi: 10.1038/nri775. [DOI] [PubMed] [Google Scholar]
- 210.Pan G, O’Rourke K, Chinnaiyan AM, Gentz R, Ebner R. The Receptor for the Cytotoxic Ligand TRAIL. Science. 1997;276:111–3. doi: 10.1126/science.276.5309.111. [DOI] [PubMed] [Google Scholar]
- 211.Chicheportiche Y, Bourdon P, Xu H, Hsu Y, Scott H, Hession C, Garcia I, Browning J. TWEAK, a new secreted ligand in the tumor necrosis factor family that weakly induces apoptosis. J Biol Chem. 1997;272:32401–4210. doi: 10.1074/jbc.272.51.32401. [DOI (VSports注册入口)] [PubMed] [Google Scholar]
- 212.Jiang S, Herrera O, Lechler RI. New spectrum of allorecognition pathways: implications for graft rejection and transplantation tolerance. Curr Opin Immunol. 2004;16:550–7. doi: 10.1016/j.coi.2004.07.011. [DOI] [PubMed] [Google Scholar]
- 213.Jiang Z, Podack E, Levy R. Donor T cells which cannot mediate perforin-dependent and FasL-dependent cytotoxicity can effect graft vs host reactivity following allogeneic bone marrow transplantation. Periodicum Biologorum. 1998;100:477–81. [Google Scholar]
- 214.Maeda Y, Levy RB, Reddy P, Liu C, Clouthier SG, Teshima T, Ferrara JL. Both perforin and Fas ligand are required for the regulation of alloreactive CD8+ T cells during acute graft-versus-host disease. Blood. 2005;105:2023–7. doi: 10.1182/blood-2004-08-3036. [DOI] [PubMed] [Google Scholar]
- 215.Voskoboinik I, Smyth MJ, Trapani JA. Perforin-mediated target-cell death and immune homeostasis. Nat Rev Immunol. 2006;6:940–52. doi: 10.1038/nri1983. [DOI] [PubMed] [Google Scholar]
- 216.Chinnaiyan A, O’Rourke K, Yu G, Lyons R, Garg M, Duan D, Xing L, Gentz R, Ni J, Dixit V. Signal transduction by DR3, a death domain-containing receptor related to TNFR-1 and CD95. Science. 1996;274:990–2. doi: 10.1126/science.274.5289.990. [DOI] [PubMed] [Google Scholar]
- 217.Krammer PH. CD95’s deadly mission in the immune system. Nature. 2000;407:789–95. doi: 10.1038/35037728. [DOI] [PubMed] [Google Scholar]
- 218.Baker MB, Altman NH, Podack ER, Levy RB. The role of cell-mediated cytotoxicity in acute GVHD after MHC-matched allogeneic bone marrow transplantation in mice. J Exp Med. 1996;183:2645–56. doi: 10.1084/jem.183.6.2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Braun YM, Lowin B, French L, Acha-Orbea H, Tschopp J. Cytotoxic T cells deficient in both functional Fas ligand and perforin show residual cytolytic activity yet lose their capacity to induce lethal acute graft-versus-host disease. J Exp Med. 1996;183:657–61. doi: 10.1084/jem.183.2.657. [DOI (VSports手机版)] [PMC free article] [PubMed] [Google Scholar]
- 220.Baker MB, Riley RL, Podack ER, Levy RB. GVHD-associated lymphoid hypoplasia and B cell dysfunction is dependent upon donor T cell-mediated Fas-ligand function, but not perforin function. Proc Natl Acad Sci USA. 1997;94:1366–71. doi: 10.1073/pnas.94.4.1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Graubert TA, Russell JH, Ley T. The role of granzyme B in murine models of acute graft-versus-host disease and graft rejection. Blood. 1996;87:1232–7. [PubMed] [Google Scholar]
- 222.Graubert TA, DiPersio JF, Russell JH, Ley TJ. Perforin/granzyme-dependent and independent mechanisms are both important for the development of graft-versus-host disease after murine bone marrow transplantation. J Clin Invest. 1997;100:904–11. doi: 10.1172/JCI119606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Ueno Y, Ishii M, Yahagi K, Mano Y, Kisara N, Nakamura N, Shimosegawa T, Toyota T, Nagata S. Fas-mediated cholangiopathy in the murine model of graft versus host disease. Hepatology. 2000;31:966–74. doi: 10.1053/he.2000.5764. [DOI] [PubMed] [Google Scholar]
- 224.Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3:745–56. doi: 10.1038/nri1184. [DOI] [PubMed] [Google Scholar]
- 225.Via C, Nguyen P, Shustov A, Drappa J, Elkon K. A major role for the Fas pathway in acute graft-versus-host disease. J Immunol. 1996;157:5387–93. [PubMed (VSports手机版)] [Google Scholar]
- 226.Hattori K, Hirano T, Miyajima H, Yamakawa N, Tateno M, Oshimi K, Kayagaki N, Yagita H, Okumura K. Differential effects of anti-Fas ligand and anti-tumor necrosis factor alpha antibodies on acute graft-versus-host disease pathologies. Blood. 1998;91:4051–5. ["V体育官网" PubMed] [Google Scholar]
- 227.van den Brink M, Moore E, Horndasch E, Crawford J, Hoffman J, Murphy G, Burakoff S. Fas-Deficient lpr Mice are More Susceptible to Graft-versus-Host Disease. Journal of Immunology. 2000;164:469–80. doi: 10.4049/jimmunol.164.1.469. [DOI] [PubMed] [Google Scholar]
- 228.Lin T, Brunner T, Tietz B, Madsen J, Bonfoco E, Reaves M, Huflejt M, Green DR. Fas ligand- mediated killing by intestinal intraepithelial lymphocytes. Participation in intestinal graft-versus-host disease. J Clin Invest. 1998;101:570–7. doi: 10.1172/JCI896. [DOI (V体育安卓版)] [PMC free article] [PubMed] [Google Scholar]
- 229.Liem LM, van Lopik T, van Nieuwenhuijze AEM, van Houwelingen HC, Aarden L, Goulmy E. Soluble fas levels in sera of bone marrow transplantation recipients are increased during acute graft-versus-host disease but not during infections. Blood. 1998;91:1464–8. [PubMed] [Google Scholar]
- 230.Das H, Imoto S, Murayama T, Kajimoto K, Sugimoto T, Isobe T, Nakagawa T, Nishimura R, Koizumi T. Levels of soluble FasL and FasL gene expression during the development of graft-versus-host disease in DLT-treated patients. Br J Haematol. 1999;104:795–800. doi: 10.1046/j.1365-2141.1999.01246.x. [DOI (VSports手机版)] [PubMed] [Google Scholar]
- 231.Martin PJ, Akatsuka Y, Hahne M, Sale G. Involvement of donor T-cell cytotoxic effector mechanisms in preventing allogeneic marrow graft rejection. Blood. 1998;92:2177–81. [PubMed] [Google Scholar]
- 232.Zimmerman Z, Shatry A, Deyev V, Podack E, Mammolenti M, Blazar BR, Yagita H, Levy RB. 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 Transplant. 2005;11:576–86. doi: 10.1016/j.bbmt.2005.05.006. [DOI] [PubMed] [Google Scholar]
- 233.Schmaltz C, Alpdogan O, Kappel BJ, Muriglan SJ, Rotolo JA, Ongchin J, Willis LM, Greenberg AS, Eng JM, Crawford JM, Murphy GF, Yagita H, Walczak H, Peschon JJ, Van Den Brink MR. T cells require TRAIL for optimal graft-versus-tumor activity. Nat Med. 2002;8:1433–7. doi: 10.1038/nm1202-797. [DOI] [PubMed] [Google Scholar]
- 234.Sato K, Nakaoka T, Yamashita N, Yagita H, Kawasaki H, Morimoto C, Baba M, Matsuyama T. TRAIL-transduced dendritic cells protect mice from acute graft-versus-host disease and leukemia relapse. J Immunol. 2005;174:4025–33. doi: 10.4049/jimmunol.174.7.4025. [DOI] [PubMed] [Google Scholar]
- 235.Brown GR, Lee E, Thiele DL. TNF-TNFR2 interactions are critical for the development of intestinal graft-versus-host disease in MHC class II-disparate (C57BL/6J-->C57BL/6J x bm12)F1 mice. J Immunol. 2002;168:3065–71. doi: 10.4049/jimmunol.168.6.3065. [DOI] [PubMed] [Google Scholar]
- 236.Brown GR, Thiele DL. Enhancement of MHC class I-stimulated alloresponses by TNF/TNF receptor (TNFR)1 interactions and of MHC class II-stimulated alloresponses by TNF/TNFR2 interactions. Eur J Immunol. 2000;30:2900–7. doi: 10.1002/1521-4141(200010)30:10<2900::AID-IMMU2900>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 237.Brown GR, Lee EL, El-Hayek J, Kintner K, Luck C. IL-12-independent LIGHT signaling enhances MHC class II disparate CD4+ T cell alloproliferation, IFN-gamma responses, and intestinal graft-versus-host disease. J Immunol. 2005;174:4688–95. doi: 10.4049/jimmunol.174.8.4688. [DOI (VSports)] [PubMed] [Google Scholar]
- 238.Xu Y, Flies AS, Flies DB, Zhu G, Anand S, Flies SJ, Xu H, Anders RA, Hancock WW, Chen L, Tamada K. Selective targeting of the LIGHT-HVEM co-stimulatory system for the treatment of graft-versus-host disease. Blood. 2006 doi: 10.1182/blood-2006-09-047332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Zimmerman Z, Jones M, Shatry A, Komatsu M, Mammolenti M, Levy R. Cytolytic pathways used by effector cells derived from recipient naive and memory T cells and natural killer cells in resistance to allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2005;11:957–71. doi: 10.1016/j.bbmt.2005.07.006. [DOI] [PubMed] [Google Scholar]
- 240.Ferrara JL, Levy R, Chao NJ. Pathophysiologic mechanisms of acute graft-vs.-host disease. Biol Blood Marrow Transplant. 1999;5:347–56. doi: 10.1016/s1083-8791(99)70011-x. [DOI] [PubMed] [Google Scholar]
- 241.Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5:987–95. doi: 10.1038/ni1112. [DOI (V体育2025版)] [PubMed] [Google Scholar]
- 242.Hill GR, Ferrara JL. The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 2000;95:2754–9. [V体育官网入口 - PubMed] [Google Scholar]
- 243.Fritz JH, Ferrero RL, Philpott DJ, Girardin SE. Nod-like proteins in immunity, inflammation and disease. Nat Immunol. 2006;7:1250–7. doi: 10.1038/ni1412. ["V体育ios版" DOI] [PubMed] [Google Scholar]
- 244.Dickinson AM, Middleton PG, Rocha V, Gluckman E, Holler E. Genetic polymorphisms predicting the outcome of bone marrow transplants. Br J Haematol. 2004;127:479–90. doi: 10.1111/j.1365-2141.2004.05216.x. [DOI] [PubMed] [Google Scholar]
- 245.Lorenz E, Schwartz DA, Martin PJ, Gooley T, Lin MT, Chien JW, Hansen JA, Clark JG. Association of TLR4 mutations and the risk for acute GVHD after HLA-matched-sibling hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2001;7:384–7. doi: 10.1053/bbmt.2001.v7.pm11529488. [DOI] [PubMed] [Google Scholar]
- 246.Nestel FP, Price KS, Seemayer TA, Lapp WS. Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor alpha during graft-versus-host disease. J Exp Med. 1992;175:405–13. doi: 10.1084/jem.175.2.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Hill GR, Teshima T, Gerbitz A, Pan L, Cooke KR, Brinson YS, Crawford JM, Ferrara JL. Differential roles of IL-1 and TNF-alpha on graft-versus-host disease and graft versus leukemia. J Clin Invest. 1999;104:459–67. doi: 10.1172/JCI6896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Krenger W, Falzarano G, Delmonte J, Snyder KM, Byon JCH, Ferrara JLM. Interferon-g suppresses T-cell proliferation to mitogen via the nitric oxide pathway during experimental acute graft-versus-host disease. Blood. 1996;88:1113–21. ["VSports在线直播" PubMed] [Google Scholar]
- 249.Piguet PF, Grau GE, Allet B, Vassalli PJ. Tumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft-versus-host disease. J Exp Med. 1987;166:1280–9. doi: 10.1084/jem.166.5.1280. ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Hill GR, Teshima T, Rebel VI, Krijanovski OI, Cooke KR, Brinson YS, Ferrara JL. The p55 TNF-alpha receptor plays a critical role in T cell alloreactivity. J Immunol. 2000;164:656–63. doi: 10.4049/jimmunol.164.2.656. [DOI] [PubMed] [Google Scholar]
- 251.Piguet PF. Tumor necrosis factor and graft-versus-host disease. In: Burakoff SJ, Deeg HJ, Ferrara JLM, Atkinson K, editors. Graft-vs-Host Disease. New York: Marcel Dekker Inc.; 1990. pp. 255–76. [Google Scholar]
- 252.Tanaka J, Imamura M, Kasai M, Masauzi N, Watanabe M, Matsuura A, Morii K, Kiyama Y, Naohara T, Higa T, Honke K, Gasa S, Sakurada K, Miyazaki T. Rapid analysis of tumor necrosis factor-alpha mRNA expression during venooclusive disease of the liver after allogeneic bone marrow transplantation. Transplant. 1993;55:430–2. [VSports在线直播 - PubMed] [Google Scholar]
- 253.Tanaka J, Imamura M, Kasai M, Masauzi N, Matsuura A, Ohizumi H, Morii K, Kiyama Y, Naohara T, Saitho M, et al. Cytokine gene expression in peripheral blood mononuclear cells during graft-versus-host disease after allogeneic bone marrow transplantation. British Journal of Haematology. 1993;85:558–65. doi: 10.1111/j.1365-2141.1993.tb03348.x. ["VSports最新版本" DOI] [PubMed] [Google Scholar]
- 254.Holler E, Kolb HJ, Moller A, Kempeni J, Lisenfeld S, Pechumer H, Lehmacher W, Ruckdeschel G, Gleixner B, Riedner C, Ledderose G, Brehm G, Mittermuller J, Wilmanns W. Increased serum levels of tumor necrosis factor alpha precede major complications of bone marrow transplantation. Blood. 1990;75:1011–6. [PubMed] [Google Scholar]
- 255.Holler E, Kolb HJ, Hintermeier-Knabe R, Mittermuller J, Thierfelde S, Kaul M, Wilmanns W. The role of tumor necrosis factor alpha in acute graft-versus-host disease and complications following allogeneic bone marrow transplantation. Transplant Proc. 1993;25:1234–6. [PubMed (VSports)] [Google Scholar]
- 256.Herve P, Flesch M, Tiberghien P, Wijdenes J, Racadot E, Bordigoni P, Plouvier E, Stephan JL, Bourdeau H, Holler E, Lioure B, Roche C, Vilmer E, Demeocq F, Kuentz M, Cahn YJ. Phase I-II trial of a monoclonal anti-tumor necrosis factor alpha antibody for the treatment of refractory severe acute graft-versus-host disease. Blood. 1992;81:1993–9. [PubMed] [Google Scholar]
- 257.Uberti JP, Ayash L, Ratanatharathorn V, Silver S, Reynolds C, Becker M, Reddy P, Cooke KR, Yanik G, Whitfield J, Jones D, Hutchinson R, Braun T, Ferrara JL, Levine JE. Pilot trial on the use of etanercept and methylprednisolone as primary treatment for acute graft-versus-host disease. Biol Blood Marrow Transplant. 2005;11:680–7. doi: 10.1016/j.bbmt.2005.05.009. ["VSports手机版" DOI] [PubMed] [Google Scholar]
- 258.Antin JH, Ferrara JLM. Cytokine dysregulation and acute graft-versus-host disease. Blood. 1992;80:2964–8. [PubMed] [Google Scholar]
- 259.Abhyankar S, Gilliland DG, Ferrara JLM. Interleukin 1 is a critical effector molecule during cytokine dysregulation in graft-versus-host disease to minor histocompatibility antigens. Transplant. 1993;56:1518–23. doi: 10.1097/00007890-199312000-00045. [DOI] [PubMed] [Google Scholar]
- 260.McCarthy PL, Abhyankar S, Neben S, Newman G, Sieff C, Thompson RC, Burakoff SJ, Ferrara JLM. Inhibition of interleukin-1 by an interleukin-1 receptor antagonist prevents graft-versus-host disease. Blood. 1991;78:1915–8. [PubMed (VSports手机版)] [Google Scholar]
- 261.Antin JH, Weisdorf D, Neuberg D, Nicklow R, Clouthier S, Lee SJ, Alyea E, McGarigle C, Blazar BR, Sonis S, Soiffer RJ, Ferrara JL. Interleukin-1 blockade does not prevent acute graft-versus-host disease: results of a randomized, double-blind, placebo-controlled trial of interleukin-1 receptor antagonist in allogeneic bone marrow transplantation. Blood. 2002;100:3479–82. doi: 10.1182/blood-2002-03-0985. [DOI] [PubMed] [Google Scholar]
- 262.Falzarano G, Krenger W, Snyder KM, Delmonte J, Karandikar M, Ferrara JLM. Suppression of B cell proliferation to lipopolysaccharide is mediated through induction of the nitric oxide pathway by tumor necrosis factor-a in mice with acute graft-versus-host disease. Blood. 1996;87:2853–60. [PubMed] [Google Scholar]
- 263.Nestel FP, Greene RN, Kichian K, Ponka P, Lapp WS. Activation of macrophage cytostatic effector mechanisms during acute graft-versus-host disease: release of intracellular iron and nitric oxide-mediated cytostasis. Blood. 2000;96:1836–43. [PubMed] [Google Scholar]
- 264.Weiss G, Schwaighofer H, Herold M. Nitric oxide formation as predictive parameter for acute graft-versus-host disease after human allogeneic bone marrow transplantation. Transplantation. 1995;60:1239–44. [PubMed] [Google Scholar]
- 265.Langrehr JM, Murase N, Markus PM, Cai X, Neuhaus P, Schraut W, Simmons RL, Hoffmann RA. Nitric oxide production in host-versus-graft and graft-versus-host reactions in the rat. J Clin Invest. 1992;90:679–83. doi: 10.1172/JCI115911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Langrehr JM, Muller AR, Bergonia HA, Jacob TD, Lee TK, Schraut WH, Lancaster JR, Hoffman RA, Simmons RL. Detection of nitric oxide by electron paramagnetic resonance spectroscopy during rejection and graft-versus-host disease after small-bowel transplantation in the rat. Surgery. 1992;112:395–401. ["V体育平台登录" PubMed] [Google Scholar]
- 267.Bogdan C. Nitric oxide and the immune response. Nat Immunol. 2001;2:907–16. doi: 10.1038/ni1001-907. [DOI] [PubMed] [Google Scholar]
