Abstract
Organ transplantation appears today to be the best alternative to replace the loss of vital organs induced by various diseases. Transplants can, however, also be rejected by the recipient. In this review, we provide an overview of the mechanisms and the cells/molecules involved in acute and chronic rejections. T cells and B cells mainly control the antigen-specific rejection and act either as effector, regulatory, or memory cells. On the other hand, nonspecific cells such as endothelial cells, NK cells, macrophages, or polymorphonuclear cells are also crucial actors of transplant rejection. Last, beyond cells, the high contribution of antibodies, chemokines, and complement molecules in graft rejection is discussed in this article. The understanding of the different components involved in graft rejection is essential as some of them are used in the clinic as biomarkers to detect and quantify the level of rejection.
Numerous cells and molecules are involved in organ rejections. Some are involved in the nonspecific innate response (e.g., macrophages); others are involved in the donor-specific adaptive response (e.g., T cells).
DIFFERENT TYPES OF REJECTION
Several types of rejection of vascularized organs can be defined according to their underlying mechanisms and tempos, the major types being hyperacute, acute, and chronic rejection. In allogeneic context and in the absence of preformed antidonor antibodies, cells and tissues are mainly rejected by acute cellular rejection mechanisms.
Hyperacute rejection appears in the first minutes following transplantation and occurs only in vascularized grafts. This very fast rejection is characterized by vessels thrombosis leading to graft necrosis. Hyperacute rejection is caused by the presence of antidonor antibodies existing in the recipient before transplantation. These antibodies induce both complement activation and stimulation of endothelial cells to secrete Von Willebrand procoagulant factor, resulting in platelet adhesion and aggregation. The result of these series of reactions is the generation of intravascular thrombosis leading to lesion formation and ultimately to graft loss. Today, this type of rejection is avoided in most cases by checking for ABO compatibility and by excluding the presence of antidonor human leukocyte antigen (HLA) antibodies by cross-match techniques between donor graft cells and recipient sera. This type of rejection is also observed in models of xenotransplantation of vascularized organs between phylogenetically distant species when no immunosuppressive treatment is given to the recipients.
Acute rejection is caused by an immune response directed against the graft and occurs between 1 week and several months after transplantation. Acute rejection is diagnosed on histological analysis of a graft biopsy according to an international classification system, the Banff classification for the kidney (Mengel et al. 2012). Acute rejection is thought to result from two immunological mechanisms that may act alone or in combination: (1) a T-cell-dependent process that corresponds to acute cellular rejection, and (2) a B-cell-dependent process that generates the acute humoral rejection. With current immunosuppressive treatment, acute rejection occurs in less than 15% of the transplants (Port et al. 2004) in nonsensitized patients.
Chronic rejection, on the other hand, is now the leading cause of graft rejection. Chronic rejection can be mediated by either humoral or cellular mechanisms linked to memory/plasma cells and antibodies. The presence of tertiary lymphoid organs in the graft is a characteristic of this form of rejection.
INNATE AND ADAPTIVE IMMUNE RESPONSES
Two major immunological mechanisms occur during allograft rejection: the nonspecific innate response that predominates in the early phase of the immune response, and the donor-specific adaptive response that results from alloantigen recognition by host T cells.
The Innate Response and Allograft Rejection
Although the adaptive response plays a central role in the mechanisms of allograft rejection, early proinflammatory signals (arising before the initiation of the T-cell response) are also considered as important factors of graft rejection. Inflammation is caused by the innate immune response induced independently of the adaptive response (Christopher et al. 2002; He et al. 2002, 2003; Land 2005). In fact, it was shown that 1 day after a heart transplant, the expression of genes coding for molecules linked to inflammation (proinflammatory cytokines, chemokines, components of the cellular infiltrate) was similar in normal mice and in mice deficient for T and B cells, but with normal NK and myeloid compartments (Rag1 or Rag2 knock-out mice) (He et al. 2003). These investigators also showed that the innate response is antigen independent, develops early after transplantation, and conditions the development of the adaptive response (He et al. 2003).
Innate immune responses are the consequence of several events associated with clinical transplantation, such as ischemia-reperfusion injury and infections, and lead to the release of damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) (Chong and Alegre 2012). DAMPs and PAMPs are recognized by so-called pattern-recognition receptors (PRRs) expressed by hematopoietic cells.
The specificity of PRRs is genetically determined and several subgroups can be classified based on their structure. The transmembrane group of PRRs includes several families of molecules such as toll-like receptors (TLRs), C-type lectins, RAGE (receptor for advanced glycation endproducts), complement receptors, scavenger receptors, and mannose receptors. The cytosolic PRR group includes retinoic acid-inducible gene-I-like receptors and nucleotide-binding domain and leucine-rich repeat-containing receptors (Iwasaki and Medzhitov 2010).
Immediately after transplantation, PRR-mediated danger signals activate dendritic cells (DCs) (LaRosa et al. 2007) leading to antigen-presenting cell (APC) maturation, up-regulation of costimulatory molecules, and secretion of proinflammatory cytokines. In this context, donor APCs migrate to the T cell areas of secondary lymphoid organs and induce the activation and differentiation of alloreactive naïve T cells into effector T helper cells. These effector cells migrate into the graft where they activate macrophages and granulocytes (neutrophils, eosinophils, and basophils) that have infiltrated the graft in response to inflammatory stimuli. The latter cells contribute to lesion formation, either directly or through the production of proinflammatory cytokines and chemokines. They also help boost and maintain the adaptive immune T-cell response. Another cell type involved in innate immunity is the NK cell. In a proinflammatory context, NK cells become activated and are able to kill target cells (Pratschke et al. 2009).
The complement system plays a central role in the effector mechanisms occurring during the innate response. The three complement pathways (classical, alternative, and mannose lectin) can be activated by DAMPs as well as by the inflammatory environment that develops during ischemia reperfusion injury (Castellano et al. 2010). Moreover, the intermediate product of complement, C5a, is able to bind to APC receptors, leading to Th1 activation and development of the adaptive immunity (Zhou et al. 2000).
Initiation of the Adaptive Response and Allograft Rejection
The adaptive immune response appears later than the innate response, its main characteristic being antigen specific. The initiation of the adaptive response is made possible by the presentation of alloantigens by APCs, mainly DCs and their allorecognition by recipient T cells. Three main pathways of allorecognition are described in the literature. The first is the direct pathway, in which donor DCs present in the graft act as passenger leukocytes. In the context of a proinflammatory environment, these cells mature and migrate to secondary lymphoid organs where they prime host T cells. The maturation of these cells is induced by proinflammatory signals such as IL-1β, TNF-α, and CD40. The second is the indirect allorecognition pathway in which recipient DCs capture, process, and present alloantigens as peptides on their host MHC molecules, and then prime T cells (Ochando et al. 2006). As dendritic cells express both MHC class I and class II, donor antigens can be presented to either CD4+ or CD8+ T cells. The third pathway is that of semidirect allorecognition, characterized by the dual ability of recipient DCs: (1) to present intact donor major histocompatibility complex (MHC) molecules acquired by cell-to-cell contact or fusion with donor exosomes, and (2) to internalize and process donor MHC as peptides on recipient MHC molecules. In this pathway, alloreactive CD8+ and CD4+ T cells are stimulated by direct and indirect allorecognition (Herrera et al. 2004; Smyth et al. 2006).
Using the two-photon microscopy technique, a recent elegant study by Bousso’s group (Celli et al. 2011) provided a better understanding of the adaptive immune response by clarifying several different issues that until then had remained hypothetical. In a murine model of skin transplantation, they showed that donor dermal DCs disappeared rapidly from the graft and migrated to draining lymph nodes (LNs). However, these donor DCs were found to be dead in the secondary lymphoid organs. This is consistent with a previous study demonstrating that donor DCs are eliminated by NK cells (Garrod et al. 2010). It has been suggested that these dying cells could be a source of alloantigens for recipient APCs present in the draining LNs that stimulate T cells by the indirect pathway. Moreover, these experiments showed that host inflammatory monocytes and DCs act as antigen-transporting cells. These recipient graft-infiltrating cells indeed have the ability to reach the draining LNs and cross-prime CD8+ T cells by the indirect pathway (Celli et al. 2011).
Initiation of the innate and adaptive immune response leads to acute and chronic graft rejection. Cellular and humoral responses, but also chemokines and innate cells, are present in both types of rejection and are described in this article.
ACUTE REJECTION
The Components of Acute Rejection
Chemokines
Chemokines are chemoattractant cytokines that influence immune cell migration. In transplantation settings, chemokines act during four key phases: (1) recruitment of mononuclear cells to the site of inflammation, (2) migration of APCs to draining LNs, (3) interaction between unprimed T cells and APCs in LNs, and (4) migration of primed alloreactive T cells to the graft (Belperio and Ardehali 2008). The roles of some chemokines in allograft rejection have been investigated directly by analyzing chemokine expression by immune cells as described in Figure 1. Furthermore, the function of these chemokine receptors and ligands has been shown using knockout (KO) mice or blocking antibodies.
Figure 1.
Chemokines: Homing of immune cells and role in acute rejection. Mononuclear cells are able to migrate into the graft or into the lymphoid organs in response to chemokine gradients. Experiments on animal models of transplantation and analysis of human samples from transplant recipients allow a better understanding of the involvement of chemokines in graft rejection.
One example is that of CCR5, which was shown to be expressed by mononuclear cells, suggesting that this chemokine helps their migration to inflammatory sites. Prolongation of cardiac allograft survival was obtained in CCR5–/– mice, demonstrating a major role for this chemokine (Gao et al. 2001). However, more recently, in a similar heart transplant model, Nozaki et al. (2007) only detected a modest effect of CCR5 on graft rejection using CCR5–/– mice. Involvement of CCR5 in graft rejection was also shown in murine models of pancreatic islet transplantation (Abdi et al. 2002). More recently, in a cardiac transplantation model in rhesus monkeys, cotreatment with a CCR5 antagonist and cyclosporine A was able to prolong heart survival. This prolongation was associated with a delay in alloantibody response, a lower graft infiltration, and a decrease in alternatively activated macrophages (Li et al. 2011). In addition to the recruitment of mononuclear cells to the graft, CCR5 polarizes T cells toward a type 1 response, and promotes the trafficking of T cells to LNs and favors DC/T-cell interaction within the LNs (Amano et al. 2005). Furthermore, Y-box protein-1, a molecule that acts as a transcription regulator of CCL5, one CCR5 ligand also called RANTES, expression in T cells, and monocytes/macrophages, was shown to be up-regulated during rejection in human kidney transplant biopsies (Raffetseder et al. 2009). Contrary to these studies, in a rat model of kidney transplant tolerance following CD28 blockade, CCL5 recruited CD4+ Tregs to the graft via a graft-to-periphery CCL5 gradient (Dilek et al. 2012).
Another well-described chemokine involved in graft rejection is CXCR3. In a murine model, absence of CXCR3 in graft recipients was shown to prolong cardiac transplant survival (Hancock et al. 2000). However, as for CCR5, a more recent study obtained controversial results showing no delay in graft rejection between controls and CXCR3 KO mice (Kwun et al. 2008). Analysis of CXCR3 and its ligands, CXCL9 and CXCL10, was performed in biopsies from transplant patients undergoing acute rejection. Fahmy et al. (2003) showed an up-regulation of CXCL10 in endomyocardial biopsies, whereas an increase in CXCL10 was detected in macrophages of lung biopsies (Agostini et al. 2001). Furthermore, the number of CXCR3+ cells present in human kidney transplant biopsies was reported to increase with graft rejection and to decrease with immunosuppressive treatment, suggesting that CXCR3 might be a good target to treat rejection (Hoffmann et al. 2006). Other recent studies have highlighted the role of chemokines as biomarkers of clinical rejection and are described in Table 1.
Table 1.
Use of chemokines, cytokines, miRNA, and cells as biomarkers of graft rejection in clinical studies
Type of rejection | Molecules/cells | Transplant organ | Samples of analysis | Significance | References |
---|---|---|---|---|---|
Acute | CXCL10 | Heart | Blood, serum | Higher serum level of CXCL10 in transplanted patients versus control ones. Among transplanted patients, high expression of CXCL10 is correlated with acute rejection. |
Crescioli et al. 2009 |
C reactive protein (CRP) | Heart | Blood, serum | Significant differential expression of CRP between rejected and nonrejected heart transplant recipient. In the first year postheart transplantation, CRP was the most useful parameter for noninvasive screening of acute cellular rejection. |
Martinez-Dolz et al. 2009 | |
Granzyme B CTLA-4 IL-6 TNF-α TGF-β |
Heart | Biopsies | Up-regulation in mRNA expression of all analyzed genes but the strongest increase is seen for granzyme B, TGF-β, and TNF-α genes. Correlation between the expression analysis of these three genes and acute rejection. |
Ramsperger-Gleixner et al. 2011 | |
CD69 | Heart | Blood, T lymphocytes | Overexpression of CD69 in T lymphocytes of patients with acute rejection. Relationship between increased expression by T lymphocytes and acute rejection is statistically more pronounced for CD8+ T cells than for CD4+ T cells. Presence of CD69+/CD8+ cells exceeding 15% in patients with acute rejection of all T lymphocytes is correlated with vigorous rejection. |
Schowengerdt et al. 2000 | |
Functional markers of CD4+ T-cell subsets: Th1 Th17 FoxP3+ T cells |
Heart | Blood and biopsies | Higher mRNA expression of the functional markers in patients with acute cardiac rejection in blood and biopsies samples (but not for Th2 mRNA cytokines). Higher percentage of Th1, Th17, and FoxP3+CD4+-circulating T cells. Graft infiltration by Th1 and Th17 cells. Kinetic changes of these CD4+ T-cell subsets after cardiac allograft. |
Wang et al. 2011 | |
CCL3L1 | Liver | Blood, PBMC | Analysis of copy number variation in genes. Higher number of CCL3L1 gene in patients with acute rejection. |
Li et al. 2012 | |
Acute | CCL2 CXCL8 CCL5 CXCL9 CXCL10 |
Liver | Blood, serum | Chemokines screening using Luminex multiplex assay. Higher expression of molecules correlated with an up-regulation of NF-κB: CCL2, CCL5, and CXCL8. Higher expression of molecules correlated with T-cell immunity: CXCL9 and CXCL10. Biomarkers of early allograft dysfunction. |
Friedman et al. 2012 |
Granzyme B Perforin |
Liver | Biopsies | Analysis by immunohistochemistry.Higher frequencies of perforin and granzyme B expression in acute rejection patients. In most of the biopsies, perforin and granzyme B are simultaneously expressed. |
Ji et al. 2004 | |
Granzyme B | Liver | Biopsies | Higher granzyme B mRNA expression in the biopsies of the acute rejection group. | Krams et al. 1995 | |
Three subsets of T cells: CD4+CD7+ CD8+CD38+ CD4+CD25+ |
Liver | Biopsies and blood | Higher number of CD4+CD7+ and CD8+CD38+ cells in blood and liver of liver recipient with acute rejection. Decreased expansion of Treg cells (CD4+CD25+) in blood and liver in patients with acute rejection. |
Perrella et al. 2006 | |
Hepatocyte-derived miRNAs (HDmiRs) | Liver | Blood, serum | Serum level of HDmiRs (miR-122, miR-148a, and miR194) are elevated up to 20-fold in patients with acute rejection. HDmiRs are promising candidates as biomarkers rejection, but also as hepatic injury, after liver transplantation. |
Farid et al. 2012 | |
CCL4 | Lung | Blood, PBMC | Analysis of copy number variation in genes. Higher number of CCL4 gene in patients with acute rejection. |
Colobran et al. 2009 | |
Treg and cytotoxic markers | Lung | Bronchoalveolar lavage and blood | Higher mRNA expression for all of the markers in patients with acute rejection, but the expression is significantly increased only for CTLA-4, FoxP3, and granzyme B mRNA. But no difference have been observed in blood samples. |
Madsen et al. 2010 | |
CD4+/CD8+ T cells NK NK-like T cells B cells Tregs Invariant receptor NK-T cells (iNKT) |
Lung | Bronchoalveolar lavage fluid | Reduction of CD4:CD8 T-cell ratio in patients with acute lung rejection. Higher frequency of CD4+ T cells expressing CD38 during rejection. Proportions of NK and Treg, as a frequency of total CD3+ T cells, trended toward being reduced in rejection. Proportions of B cells, NK-like T cells, and the percentage of CD4−/CD8− iNKT cells trended toward being increased in rejection. |
Gregson et al. 2008 | |
Acute | CD4+/CD8+ T cells B cells Memory T cells |
Lung | Bronchus-associated lymphoid tissues | Reduction of CD4:CD8 T-cell ratio is correlated with elevated acute rejection episode for patients in the first year of transplantation. | Shenoy et al. 2012 |
CXCR1 CXCR2 |
Kidney | SNP genotype | Donor CXCR1-2668 GA/AA genotype is associated with acute rejection in kidney transplantation. | Ro et al. 2011 | |
CXCL10 CXCL13 |
Kidney | Blood, PBMC | Overexpression of CXCL10 and CXCL13 in PBMC of patients with acute rejection. | Mao et al. 2011 | |
Granzyme B Perforin FasL |
Kidney | Blood,T lymphocytes | Significant higher perforin mRNA expression in patients with acute rejection. No consistent pattern of granzyme B and FasL expression. |
Shin et al. 2005 | |
CD69 expression | Kidney | Blood,T lymphocytes | Higher percentages of CD69+ cells in the CD3+ and CD8+ T-cell subsets of patients with acute rejection. Correlation between CD69+CD8+ T cells and acute rejection is the more clinically useful test (based on receiver-operator characteristics). |
Posselt et al. 2003 | |
CXCL10 | Kidney | Blood, serum | High serum level of CXCL10 is associated with acute risk of graft failure. | Rotondi et al. 2004 | |
CXCL9 | Kidney | Blood, serum | High level of CXCL9 pretransplantation in recipient patients is a marker of high risk of acute rejection. | Rotondi et al. 2010 | |
Pattern of cytokines Granzyme B Perforin |
Kidney | Blood, PBMC | High level of proinflammatory cytokines (IL-4, IL-5, IL-6, and IFN-γ) and cytotoxic molecules (perforin and granzyme B) are associated with acute rejection in kidney transplant recipient. | Dugre et al. 2000 | |
CXCL9 CXCL10 |
Kidney | Urine | Increased expression of CXCL9 and CXCL10 in urine of patients with acute rejection. | Jackson et al. 2011a | |
Granzyme B Perforin |
Kidney | Urine | Increased mRNA expression of granzyme B and perforin in urine of patients with acute rejection. | Li et al. 2001 | |
Acute | Perforin Granzyme B FasL PI-9 FoxP3 |
Kidney | Urine | Increased mRNA signature expression in urine of patients with acute rejection. FoxP3 mRNA level is the most accurate and predictive for acute rejection with a sensibility and specificity of 100%. |
Aquino-Dias et al. 2008 |
miRNAs miR-210 |
Kidney | Urine | Down-regulation of miR-10b and miR-210 in patients with acute rejection (compared with controls). Up-regulation of miR-10a in patients with acute rejection (compared with controls). Expression of miR-210 is only one that changes before and after rejection episode. Low expression of miR-210 is correlated with acute rejection and prediction of long-term of kidney function. |
Lorenzen et al. 2011 | |
CXCR3 | Kidney | Biopsies | Increase number of CXCR3-positive cells in biopsies from patients with acute rejection and decrease with immunosuppressive treatment. | Hoffmann et al. 2006 | |
CX3CR1 | Kidney | Biopsies | CX3CR1 intragraft staining is associated with worst outcome. CX3CR1 is expressed by macrophage and dendritic cells in acute rejection. |
Hoffmann et al. 2010 | |
Pattern of chemokines | Kidney | Biopsies | Increase of inflammatory chemokines and chemokine receptors transcripts during acute rejection: CCL3/CCL5/CXCL9/CXCL10/CXCL11 and CCR5/CCR7/CXCR3. Rejection associated with Th1 polarization. |
Lo et al. 2011 | |
miRNAs | Kidney | Biopsies | Up-regulation of seven miRNA and down-regulation of 10 miRNA samples of patients with acute rejection. Three samples (overexpressed) are predictive with a high accurancy of acute rejection: miR-142-5p, miR-155, and miR-223. |
Anglicheau et al. 2009 | |
Chronic | CCR4 | Lung | Blood, PBMC | Reduction of CCR4+ expression on CD4+ memory T cells in patients with chronic rejection. | Paantjens et al. 2011 |
CXCL9 CXCL10 |
Lung | Bronchoalveolar lavage fluid | Cumulative expression of CXCL9 and CXCL10 in bronchoalveolar liquid is a marker of graft failure. | Neujahr et al. 2012 | |
Chronic | HNPs (human neutrophil peptides 1–3) | Lung | Bronchoalveolar lavage fluid | High level of HNPs peptides by mass spectrometry in patients who have developed bronchiolitis obliterans syndrome (marker of chronic lung rejection). Validation by enzyme-linked immunosorbent assay (ELISA). Monitoring levels of HNPs can predate the clinical onset of disease up to 15 months. |
Nelsestuen et al. 2005 |
CCL2 | Kidney | Urine | Early detection of CCL2 in urine is a marker of a development of IF/TA at 24 months. | Ho et al. 2010 | |
CXCL9 CXCL10 |
Kidney | Urine | Increased expression of CXCL9 and CXCL10 in urine of patients with chronic rejection. | Schaub et al. 2009 | |
CCR1 RANTES |
Kidney | Biopsies | CCR1 and RANTES promote ongoing IF/TA and are linked to worst outcome. | Dikow et al. 2010 | |
Immunoproteasome β subunit 10 | Kidney | Blood and biopsies | Immunoproteasome β subunit 10 mRNA expression is specifically up-regulated during chronic active antibody-mediated rejection. | Ashton-Chess et al. 2010 | |
Tribbles-1, -2, and -3 | Kidney | Blood, biopsies and urine | Only mRNA expression of Tribbles-1 is significantly increased in blood and graft biopsies of kidney transplant recipients with chronic antibody-mediated rejection compared to patients with other histological and clinical diagnoses. In peripheral blood, mononuclear cell mRNA Tribbles-1 expression discriminates patients with chronic antibody-mediated rejection from those with other types of late allograft injury with high sensitivity and specificity. Tribbles-1 was found to be a potential blood and tissue biomarker of chronic antibody-mediated rejection. |
Ashton-Chess et al. 2008 | |
Acute and chronic | CXCL10 | Kidney | Blood, serum | High serum levels of CXCL10 is associated with both acute rejection and chronic allograft nephropathy. | Lazzeri et al. 2005 |
CXCL10 MIG MIP-1 OPG |
Kidney | Urine | Chemokines screening using Luminex multiplex assay. CXCL10 and MIG: markers of acute rejection, but not chronic rejection and stable graft function. MIP-1 (Δ) and OPG: markers of both acute and chronic rejection, but not stable graft function. |
Hu et al. 2009 | |
FoxP3 T-bet Granzyme B |
Kidney | Blood, PBMC and biopsies | mRNA expression of FoxP3 and T-bet increase in biopsies, but not in PBMC samples of patients with acute rejection. mRNA expression of granzyme B increased in biopsy samples of patients with acute (cellular) and chronic (antibody-mediated) rejection. mRNA expression of granzyme B in PBMC is significantly reduced for patients with chronic rejection. Granzyme B, but not FoxP3 and T-bet, should be considered as a biomarker of chronic rejection. |
Ashton-Chess et al. 2009 |
IF/TA, interstitial fibrosis and tubular atrophy.
In mouse models of cardiac and pancreatic islet transplantation, expression of CXCR3 was detected in Tregs, memory CD4+ and CD8+ T cells, NKT, and NK cells (Uppaluri et al. 2008). Zhai et al. (2006) showed that CXCR3 is involved in the recruitment of CXCR3+CD4+ T cells following ischemia-reperfusion injury in a model of rat syngeneic orthotopic liver transplantation. Similarly, expression of CXCR3 by Tregs resulted in their recruitment to peripheral sites of inflammation, and a high number of circulating CXCR3+ Tregs was detected in kidney transplant patients treated with an mTOR inhibitor as their immunosuppressive therapy (Hoerning et al. 2011). In addition to the role of CXCL9/CXCL10 in the recruitment of T cells to the allograft, Rosenblum et al. (2010) recently showed that these chemokines regulate donor-specific CD8+ T-cell priming. As such, in a murine model of cardiac transplantation using KO mice for either the CXCL9 or CXCL10 gene, CXCL9 promoted, whereas CXCL10 inhibited the differentiation of IFN-γ-producing donor-specific CD8+ T cells.
A new molecule called TAK-779 was identified as a blocking agent for both CCR5 and CXCR3. This molecule was shown to prevent cardiac allograft rejection in mice (Akashi et al. 2005) as well as kidney rejection in rats (Kakuta et al. 2012). In the latter study, prolongation of graft survival correlated with a decrease in macrophage infiltration (Kakuta et al. 2012). Last, coexpression of CXCL10 and fractalkine or CX3CL1 by endothelial cells was shown to be involved in effector memory T-cell recruitment (Manes et al. 2007; Manes and Pober 2008). Fractalkine acts as a chemoattractant and adhesion molecule for cells that express CX3CR1, including certain immune cells such as monocytes/macrophages, NK cells, and T cells (Imai et al. 1997; Fong et al. 1998; Foussat et al. 2000).
Some studies have also been performed to analyze other chemokine receptors. For example, CCR2 was shown to be an important chemoattractant receptor for mononuclear cells. This is because CCR2 induced mobilization of monocytes from bone marrow to blood and an accumulation of CD11c+ cells in the allograft in a mouse model of lung allotransplantation (Gelman et al. 2010). Interaction of CCR2 with its ligand (CCL2) also caused T-cell clonal expansion and differentiation (Lee et al. 2003). In CCR2–/– mice, an increase in graft survival was observed after pancreatic islet transplantation (compared to control mice) and associated with the absence of CD8+ effector T cells in these mice (Abdi et al. 2004).
CCR4 is an important chemokine for homing of memory T cells (Campbell et al. 1999) and was shown to be crucial for the recruitment of FoxP3+ Tregs in a model of tolerance induced by donor-specific transfusion/CD154 therapy (46) as no tolerance was achieved in CCR4-deficient mice (Lee et al. 2005). Inhibition of CCR4 and its ligands led to an increase in heart graft survival associated with a decreased in DC/T-cell interactions and an inhibition of monocytes and NK cells present in the graft (Alferink et al. 2003; Huser et al. 2005). Last, CCR7 is known to be involved in the localization of both DCs and T cells in the T-cell-rich zones of the LNs, indicating a role for CCR7 in T-cell homing and priming (Hopken et al. 2004). Recently, Liu et al. (2011) showed that a lack of CCR7 prevented tolerance induction in a cardiac murine model of transplantation. This impairment of tolerance was partially rescued by adoptive transfer of wild-type plasmacytoid DCs to the CCR7-deficient mice (Liu et al. 2011).
As discussed before, some of these chemokines and chemokine receptors might be useful as markers of acute rejection. In the case of renal transplantation, rejection could be monitored via noninvasive urine sampling. The most well-known urine marker is IP-10 or CXCL10. An increase in IP-10 mRNA and protein in the urine of a kidney transplant recipient was shown to correlate with incidence of acute rejection episodes (Matz et al. 2006). Moreover, high levels of CXCL3 (Tatapudi et al. 2004) and CD103 mRNA (Ding et al. 2003) were present in a recipient undergoing acute rejection. Nevertheless, chemokines are not the only markers of rejection. In fact, other proteins, such as granzyme B and FoxP3, miRNAs, and cells could also be used as predictive biomarkers of rejection (Table 1).
Nonspecific Effector Cells (NK Cells, Macrophages, Monocytes, DCs, and Neutrophils)
Despite the finding that NK cell detection in the graft infiltrate was a negative prognostic factor (Sorrentino et al. 2006), this cell type does not appear to be necessary or sufficient to mediate allograft rejection. Some recent studies have even highlighted the involvement of NK cells in the phenomenon of tolerance (van der Touw et al. 2012). NK cells seem to act mainly as a bridge between innate and adaptive immune responses. This has been underlined by experiments using CD28-deficient mice. For example, by depleting NK1.1+ cells using antibodies in CD28-deficient mice, some groups showed that absence of NK cells reduce cardiac allograft rejection in mice (Maier et al. 2001; McNerney et al. 2006). In this model, NK cells, but not NKT cells, were required for rejection by promoting T-cell expansion and effector function (McNerney et al. 2006). This effect was independent of the NK-cell activator NKG2D receptor.
In contrast, several studies have highlighted the role of the NKG2D receptor in rejection. In a model of cardiac transplantation in CD28-deficient mice, blocking NKG2D prevented graft rejection (Kim et al. 2007). Zhang and coworkers confirmed the involvement of NKG2D in graft rejection as they showed that the NKG2D receptor present on NK cells recognizes renal tubular epithelial cells, leading to their killing by a perforin-dependent pathway during ischemia reperfusion injury (Zhang et al. 2008). Promotion of CD4+ T-cell activation by the indirect allorecognition pathway is another mechanism of action of NK cells investigated by NK-cell depletion and NKG2D blockade in a murine skin graft model (Ito et al. 2008). Two ligands of NKG2D, namely RAEL1 and RRLT, were found to be expressed in rat liver allograft and to be linked to acute rejection (Zhuo et al. 2010). Moreover, in human kidney transplant biopsies, an elevated level of NKG2D mRNA was also associated with acute rejection (Seiler et al. 2007). NK cells produce a variety of cytokines, including IFN-γ, TNF-α, and TGF-β, as well as colony-stimulating factors (Cuturi et al. 1987, 1989; Anegon et al. 1988). IFN-γ produced by NK cells is known to induce an up-regulation of MHC class I and class II molecules by endothelial cells leading to their killing by CD8+ T cells (McDouall et al. 1997; Ayalon et al. 1998). More recently, a role for IL-15 in NK-cell-mediated mechanisms was shown, as in vivo stimulation of NK cells with IL-15 was found to induce skin graft rejection in RAG–/– mice (Kroemer et al. 2008). Last, an interesting effect of NK cells on donor DCs has been described. Coudert et al. (2002) showed that interaction of NK cells with donor DCs in the absence of CD8+ T cells promoted CD4+ T-cell priming and regulated the Th balance. In a more recent study performed in a model of CD4+ T-cell-mediated allogeneic skin graft rejection, NK cells killed donor DCs in draining LNs by a perforin-dependent pathway that avoided T-cell activation (Laffont et al. 2008) thereby confirming that NK cells favor indirect allorecognition (Garrod et al. 2010).
Macrophages constitute another innate cell population that contributes to the tissue damage observed in rejected organs. In humans, macrophages were found to represent 40%–60% of the cellular infiltrate in renal allotransplants (Hancock et al. 1983). A large number of monocytes were also detected in infiltrates from the graft biopsies of kidney transplant recipients under T-cell-depleting therapy (anti-CD52 and FK506 monotherapy) (Salama et al. 2007). These cells were recruited to inflammatory sites by monocyte chemoattractant protein-1 (MCP-1). Blockade of MCP-1 was shown to increase pancreatic islet transplant survival in rodents (Lee et al. 2003). In addition, blockade of macrophage colony-stimulating factor-reduced macrophage proliferation and accumulation in the graft leading to a decrease in the severity of kidney transplant rejection in mice (Jose et al. 2003). Other studies have confirmed in macrophages a decreased infiltration and an increase in graft survival in murine models of both kidney (Qi et al. 2008) and heart (Takeiri et al. 2011) transplantation. Furthermore, in the context of human kidney transplantation, CX3CR1 expression by macrophages was associated with acute rejection, and was a negative prognostic factor in human kidney graft (Hoffmann et al. 2010).
There are several macrophage subsets, and specialized tissue-resident macrophages are widely distributed across the body including the liver (Kupffer cells) and bone (osteoclasts). Macrophages can also display heterogeneity in terms of their functional phenotype as well as their activation state. In the same terms, activated macrophages are classified as classically or alternatively activated macrophages, named M1 and M2 macrophages, respectively, referring to T-cell nomenclature (Mantovani et al. 2004).
M1 macrophages are activated by a combination of IFN-γ and TNF released by Th1 cells. M1 macrophages have the capacity to present antigen to T cells and express high levels of costimulatory molecules (CD86 and CD80) (Mantovani et al. 2004). These cells also express large amounts of the proinflammatory cytokine IL-12 and promote the development of the Th17 immune response by secreting IL-1, IL-6, and IL-23. M1 macrophages are thought to play a role in the defense against bacteria or viruses as well as tumor resistance. However, in several conditions, this macrophage subset can take part in chronic inflammation and autoimmune diseases (Mosser and Edwards 2008).
M2 macrophages, on the other hand, are associated with immunoregulation. Macrophage phenotype and function are thought to differ according to the chemokine environment at the time of their activation (Mantovani et al. 2004). In fact, macrophages develop a regulatory profile when activated by IL-10 or immune complexes associated with a second stimulus. These cells act as regulatory cells by producing high levels of IL-10 and other anti-inflammatory cytokines such as TGF-β, for example (Mosser and Edwards 2008). Alternatively, if resident macrophages are activated in the presence of IL-4 and IL-13, they are considered as M2a, or wound-healing macrophages, as they promote tissue repair. The latter cells are characterized by an up-regulation of mannose receptor and production of polyamine (Mantovani et al. 2004; Mosser and Edwards 2008).
However, depending on the context, activated macrophages are able to switch their phenotype and change their function too (Mosser and Edwards 2008). One example of this is the differentiation of M1 to hybrid macrophages that share the characteristics of both regulatory and wound-healing macrophages. Unfortunately, these cells are tumor-associated macrophages and inhibit antitumor immunity (Duluc et al. 2007). Another example is in the context of the autoimmune disease diabetes, in which adipose-tissue-associated macrophages with a wound-healing phenotype polarize toward a phenotype similar to classically activated macrophages (Lumeng et al. 2007).
A recent study performed in a rat model of lung transplantation showed that CD68+ macrophages were the most abundant cell type observed during acute rejection, whereas CD163+ cells were distributed around vessels and bronchioles (Jungraithmayr et al. 2010). Regarding their tissue localization, macrophage infiltration was significant in the interstitium, but also in the arterial intima of vascularized grafts (Sun et al. 2011). Infiltration was also observed in the glomeruli of kidney transplants.
In contrast to the arterial intima in which no correlation between antibody-mediated rejection (AMR) and macrophage infiltration was shown (Kozakowski et al. 2009), macrophages/monocytes were associated with AMR when present in the glomeruli. Moreover, presence of focal or diffuse C4d deposits was associated with macrophage/monocyte infiltrates, whereas T cells predominated when no C4d deposits were detected (Magil 2005; Magil and Tinckam 2006). These results were confirmed by Fahim et al. (2007), who showed an abundant recruitment of monocytes to peritubular and glomerular capillaries during humoral rejection. Moreover, in a murine model of hepatocellular allotransplantation in which rejection is mediated by a CD4-dependent alloantibody response, depletion of macrophages delayed hepatocyte rejection and inhibited in vivo allocytotoxicity (Horne et al. 2008).
Polymorphonuclear cells are also efficient producers of chemokines (Molesworth-Kenyon et al. 2005). As described in the previous section, chemokines act on T-cell activation, proliferation, and function. One type of polymorphonuclear cell, the neutrophil, was detected in large numbers among the cells infiltrating murine skin allografts during acute rejection (Celli et al. 2011). Neutrophils have also been shown to contribute to allograft rejection in models of costimulation blockade (El-Sawy et al. 2005), or in the absence of IFN-γ (Miura et al. 2003) and IL-4 (Surquin et al. 2005). There is also evidence of eosinophil involvement in models of rejection and their activation was shown to be dependent on a Th2 CD4+ T-cell response (IL-4-dependent) (Braun et al. 2000; Surquin et al. 2005). Eosinophils were also abundant in intestinal graft infiltrates on T-cell elimination (Wu et al. 2006).
Recently, the role of platelets has been shown in allograft rejection, as these cells are able to induce immune stimulation. In this context, platelets are believed to act by recruiting mononuclear cells by secreting cytokines/chemokines and by stimulating monocytes, macrophages, and T cells by interaction with them via P-selectin/PSGL-1 or CD40/CD40L pathways (Xu et al. 2006; Kirk et al. 2009).
Last, endothelial cells are also able to induce allogeneic CD4+ and CD8+ T-cell activation and proliferation, favoring IFN-γ and IL-2 secretion and leading to graft rejection (Epperson and Pober 1994). In a recent review, Taflin et al. (2011, 2012) described how in the context of inflammation, such as observed in transplantation, endothelial cells are activated by proinflammatory cytokines and TLR ligands. This activation leads to memory CD4+ T-cell proliferation and Th1/Th17 expansion by endothelial cell secretion of proinflammatory cytokines and chemokines (such as CXCL10, IL-6, and IL-1α) and overexpression of costimulatory molecules (such as LFA-3 or OX-40L) or adhesion molecules (such as ICAM-1) (Griffin et al. 2012). In humans, three nonclassical MHC class I antigens exist: HLA-E, HLA-F, and HLA-G. Coupel et al. (2007) showed that HLA-E is expressed only by leukocytes, such as NK cells, B and T lymphocytes, macrophages, and by endothelial cells. Moreover, they show that endothelial cells up-regulate HLA-E in inflammatory conditions, and endothelial cells produce a soluble form of HLA-E. Coupel’s group additionally showed that increased expression of the membrane-bound form of HLA-E protected IFN-γ-activated endothelial cells from CD94/NKG2A-mediated cytolysis, whereas the soluble form of HLA-E protected other cell types. Endothelial cells expressed the MHC class-I related chain A, MICA, which is a ligand for NKG2D. NK cells and cytotoxic T cells express this activating receptor. Under inflammatory stimulation by TNF-α, human endothelial cells up-regulate the expression of MICA (Lin et al. 2012). These results are consistent with previous data suggesting that MICA is expressed by kidney macrovascular endothelial cells in kidney transplant recipients (Sumitran-Holgersson et al. 2002). Moreover, this study showed that MICA acts as an antibody target, which confirmed anti-MICA antibodies are associated with the reduction of graft survival (Zou et al. 2007).
Acute Cellular Response
T-Cell Activation and Effector T Cells
Effector T cells can mediate cell lysis either via cytokine or chemokine secretion, which induces necrosis of transplant tissue, or via direct contact with epithelial or endothelial cells and various mechanisms of cytotoxicity. However, T cells can also stimulate B cells to initiate humoral rejection, or other cells such as macrophages or neutrophils, whose role in transplant rejection is described above.
On activation by DCs, naïve alloreactive CD4+ T cells can differentiate into T helper cells, including Th1, Th17, Th2, or into regulatory T cells (Tregs). In a proinflammatory environment, naïve CD4+ T cells differentiate mainly into Th1 and Th17 cells. Th1 cells produce IFN-γ and IL-2 and are involved in cytotoxic T lymphocyte (CTL) priming, stimulation of the humoral response, and activation of other cell types such as NK cells. IL-17 produced by Th17 cells stimulates the production of inflammatory cytokines and chemokines leading to the recruitment of neutrophils and macrophages to the graft. In a Th1-depleted environment, Th17 cells (Yuan et al. 2008) or Th2 cells (Barbara et al. 2000) can efficiently promote graft rejection. Th2 cells release anti-inflammatory cytokines such as IL-10 and IL-4 T follicular helper cells and Th9 helper cells have not yet been described in detail, but they have been shown to induce B-cell maturation and mast cells recruitment, respectively.
In contrast, CD4+ Tregs have been shown to be involved in the tolerance process and to prevent graft rejection in mice (Kingsley et al. 2002). These cells are able to suppress CD4+ and CD8+ effector T cells and can also target APCs, decreasing their capacity for antigen-presentation and costimulation (Shevach 2009). A recent study performed using humanized mice showed the superiority of donor alloantigen-specific CD4+ Tregs over their polyclonal counterparts to suppress alloimmune responses (Sagoo et al. 2011). In tolerant liver transplant patients, an increase in peripheral blood CD4+ Tregs has been observed by several groups (Li et al. 2004; Martinez-Llordella et al. 2007; Pons et al. 2008; Nafady-Hego et al. 2010). Although this Treg signature does not seem to be present in the peripheral blood of kidney transplant patients, an increase in intragraft Tregs was detected in such patients (Bestard et al. 2007).
CD8+ T cells can differentiate into CTLs, which themselves can be classified as Tc1, Tc2, or the more recently described Tc17 (Yuan et al. 2009). Although, naïve CD8+ cells can only be activated by DCs, CD4+ T cells have been shown to facilitate CD8+ T-cell differentiation either by cell-to-cell contact or by secretion of IL-2/IFN-γ cytokines. In the latter case, both APCs and CD4+ helper T cells have been shown to be involved in CTL differentiation (Ridge et al. 1998). CTLs subsequently migrate to the graft where they recognize target cells via their allogeneic MHC class I molecules. Killing by CTLs is mediated mainly by the secretion of perforin and granzyme B, or by the Fas/FasL pathway. These two pathways induce target cell apoptosis. CTLs are also able to secrete soluble mediators such as TNF-α. In terms of the dynamics of CTL action, in a model of skin graft rejection, an accumulation of CTLs surrounding the graft was detected, followed by an early phase of killing in small areas of the dermis/epidermis junction. The CTLs were shown to disseminate to the whole graft and induce tissue destruction (Celli et al. 2011).
Compared to their naïve counterparts, memory T cells induce a faster and more effective immune response with lower antigen stimulation (Rogers et al. 2000; Veiga-Fernandes et al. 2000). This is supported by data in humans showing that the pretransplant frequency of donor-specific memory cells correlates with the posttransplant risk of developing acute rejection episodes (Heeger et al. 1999). These results were confirmed in liver transplant patients in whom the presence of a high number of CD8+ cells with a memory phenotype before transplantation was associated with reduced graft survival (Tanaka et al. 2006). More recently, in a model of kidney allotransplantation in nonhuman primates, Nadazdin et al. (2011) showed that the presence of memory T cells before transplantation was a barrier to transplantation tolerance.
Despite CD8+ Tregs being of considerable interest in transplantation, their subsets and mechanisms of action are less understood in comparison to their CD4+ Treg counterparts (Guillonneau et al. 2010). Understanding the function and mechanisms of action of the different CD8+ Treg subsets is essential. In rodent models, several studies have shown the role of CD8+ Treg subsets in the suppression of transplant rejection. For example, donor-antigen-specific CD8+CD45low Tregs were shown to develop in a fully mismatched heart allograft model in rat on treatment with CD40Ig (Guillonneau et al. 2007). The therapeutic potential of these regulatory cells has also been highlighted in human studies; in liver and cardiac transplant recipients, expansion of CD8+CD28− Tregs was shown to correlate with low exposure to immunosuppressive drugs and a decrease in acute and chronic allograft rejection (Guillonneau et al. 2010).
Memory T Cells
The presence of memory T cells able to react with alloantigens was shown in healthy individuals by Lombardi’s group in 1990 (Lombardi et al. 1990). Memory T cells showed a faster proliferation, in accordance with observations in vivo during secondary-type immune responses (Akbar et al. 1990). Memory CD8+ T cells act as CTLs as they are able to directly kill target cells, mainly via the granzyme/perforin pathway (Barber et al. 2003).
Two distinct subsets of memory T cells have been identified, and are termed central and effector memory cells (Brook et al. 2006). Central and effector memory cells are CD45RO+ CD45RA−, but differ by their expression of the CD62L/CCR7 markers. Contrary to central memory cells, effector memory cells express more β1 and β2 integrins. Moreover, the two subsets differ in their localization because central memory T cells express intermediate levels of CCR4 and CCR6, whereas effector memory T cells express high levels of CCR1, CCR3, and CCR5. Although central memory T cells require restimulation, this subset is more efficient in their killing function (Sallusto et al. 1999; Barber et al. 2003; Hengel et al. 2003).
In sensitized patients, the presence of memory T cells specific for alloantigens can be caused by prior blood transfusion, pregnancy, or transplant rejection. The presence of allospecific memory T cells is more difficult to explain in nonsensitized individuals, but may be the result of heterologous immunity or homeostatic proliferation.
Heterologous immunity is defined as the ability of alloreactive memory T cells specific for a microbial antigen to cross-react with allogeneic MHC molecules through direct allorecognition. This affinity of the T-cell receptor for allogeneic MHC molecules can be higher than that for self-antigens. Heterologous immunity has been shown both in mice and in humans (Smith et al. 2012). Homeostatic proliferation occurs in lymphopenic conditions (such as those caused by certain immunotherapies) and in the absence of an antigenic stimulus. In these conditions, peripheral T cells rapidly proliferate and acquire a memory phenotype that spontaneously skews toward a Th1 phenotype (Moxham et al. 2008). Certain cytokines, such as IL-7, have been shown to mediate this homeostatic proliferation (Schluns et al. 2000).
Memory T cells generated in the context of viral infections or by homeostatic proliferation may prevent tolerance induction, as shown using well-established rodent models of tolerance (Valujskikh et al. 2002; Zhai et al. 2002; Wu et al. 2004). These results show that memory T cells can act as barriers to tolerance induction. It is therefore necessary to develop therapies to prevent memory T-cell generation or to eliminate such cells. The effect of immunosuppressive drugs on memory T cells has started to be studied. So far, it is known that CD4+ memory T cells are resistant to steroids, deoxyspergualin, and sirolimus. In contrast, calcineurin inhibitors tacrolimus and cyclosporine A inhibit their proliferation and in vitro activation (Pearl et al. 2005). Optimal activation of memory T cells requires costimulatory molecules, which are different from those acting on naïve cells, such as ICOS/ICOSL, OX40/OX40L, and CD27/CD70 (Croft 2003; Wu et al. 2004). Consequently, blockade of the CD27/CD70 pathway prolongs the survival of heart transplant in mice (Yamada et al. 2005). Moreover, IL-7 inhibition blocks allograft rejection mediated by memory T cells without affecting Tregs (Wang et al. 2006).
Acute Humoral Response
Although T cells have been considered for many years as playing a predominant role in graft rejection, it is now known that the acute humoral response is the main cause of acute graft loss (Terasaki 2003). In fact, acute humoral rejection accounts for 15% to 20% of graft rejection within the first posttransplant year, despite immunosuppressive therapies (Montgomery et al. 2004; Lucas et al. 2011).
The criteria for the diagnosis of acute humoral rejection of kidney transplants were initially established by the Banff working group in 2003 (Racusen et al. 2003). At least one of the following criteria must be present for a diagnosis of acute humoral rejection to be made: (1) morphological criteria: presence of neutrophils and/or monocytes/macrophages in peritubular capillaries (PTC) and/or glomeruli, fibrinous arterial necrosis, thrombosis in the glomerular capillaries, arterioles, and/or small arteries and acute tubular damage; (2) immunohistological criteria: C4d deposits in PTC, presence of immunoglobulins, or complement in the fibrinous necrosis of the arteries; (3) serological criteria: presence of circulating antidonor (HLA and non-HLA) antibodies called donor-specific antibodies (DSA).
These criteria were optimized during the 9th Banff conference by including the quantification of C4d deposition (Solez et al. 2008).
Generally, detection of DSA and C4d deposits is associated with an increase in AMR and an inferior graft outcome (Kedainis et al. 2009). Nevertheless, although the presence of C4d deposits in kidney biopsies appears to be a good marker of AMR, during the last Banff meeting it was agreed that AMR can also be diagnosed in the absence of C4d detection (Mengel et al. 2012).
The new objectives of the Banff committee are therefore to characterize AMR by defining: (1) thresholds for microvascular injury and for DSA in AMR, noting that all injuries are specific and all patients with DSA develop AMR, (2) C4d-negative AMR before adding this category to the Banff classification, (3) acute versus chronic AMR, and (4) the significance of intimal arteritis in the absence of necrosis as a histological criteria (Mengel et al. 2012).
B Cells
The importance of B cells in allograft rejection was shown using B-cell-deficient mice. Several groups, including Brandle and coworkers, as well as Wasowska and colleagues showed that acute allograft rejection was delayed in B-cell-deficient mice and could be restored by the administration of hyperimmune sera (Brandle et al. 1998; Wasowska et al. 2001). In humans, the primary role of B cells as amplifiers of the alloresponse is suggested by the positive reports of B-cell depletion therapy (rituximab) as induction for ABO-incompatible renal transplant recipients (Tyden et al. 2009). However, rituximab-induction therapy was reported to lead to acute cellular rejection in some kidney transplant recipients (Clatworthy et al. 2009). It is therefore thought that B-cell depletion may be quicker at depleting the B cells that contribute to tolerance than those that promote rejection, thereby temporarily amplifying the alloresponse.
The ability of B cells to present antigen and to produce antibodies and cytokines means that these cells can take part in allograft rejection by different mechanisms. B cells have the ability to promote T-cell activation/differentiation through costimulatory pathways, or cytokine release (Constant 1999; Zarkhin et al. 2010). B cells have also been shown to promote alloreactive T-cell differentiation into memory T cells (Ng et al. 2010). Moreover, a deficiency in B-cell antigen presentation can prolong graft survival (Noorchashm et al. 2006).
More recently, several groups suggested that B cells might be involved in tolerance. For example, Deng and coworkers showed that anti-CD45RB-induced tolerance to heart transplants requires the presence of B cells (Deng et al. 2007). Furthermore, resting B cells expressing donor antigen indefinitely prolonged heterotopic heart transplant survival (Niimi et al. 1998). In rats, long-term allograft tolerance is characterized by an accumulation of B cells expressing genes such as BANK-1, which are associated with tolerance (Le Texier et al. 2011). In addition, anti-CD20-mediated depletion of B cells in mice was shown to accelerate skin graft rejection (DiLillo et al. 2011). A specific population of B cells expressing TIM-1 was identified as having regulatory properties in mouse models of islet transplantation (Ding et al. 2011). Furthermore, a B-cell signature was described in immunosuppressant-free transplant patients who were spontaneously tolerant to their HLA mismatched kidney transplant (Newell et al. 2010; Pallier et al. 2010; Sagoo et al. 2010; Silva et al. 2012). In contrast, spontaneously tolerant liver transplant recipients were reported to display an NK cell signature (Martinez-Llordella et al. 2008). These results were confirmed by the analysis of PBMC from kidney and liver transplant patients by the same laboratory, showing that the B-cell signature was only detectable in kidney in immunosuppressant-free kidney transplant patients (Lozano et al. 2011).
Antibodies
Halloran’s group showed the role of alloantibodies in acute humoral rejection in the 1990s. They described a correlation between the prognosis of acute rejection and the production of antidonor antibodies after transplantation (Halloran et al. 1990). In presensitized patients, the presence of DSA at the time of transplantation was identified as a high risk factor for AMR and patients who develop anti-HLA DSA tend to have inferior long-term graft survival compared to those that do not (Lefaucheur et al. 2008, 2010). In support of these results, a recent study by Cooper et al. (2011) also showed that patients who develop de novo DSA after transplantation have an inferior graft outcome.
Alloantibodies that develop against the donor organ can recognize several types of antigens (for review, see Dragun 2008): HLA antigens class I and II (Terasaki and Ozawa 2004; Terasaki and Cai 2005), MICA and MICB antigens (MHC class I-related molecules A and B) (Zou et al. 2007; Li et al. 2010), minor histocompatibility antigens and non-HLA antigens including the angiotensin II type 1 receptor (Dragun et al. 2005), vimentin (Mahesh et al. 2007), myosin, the ABO blood group antigens (Montgomery et al. 2012), perlecan, type IV and VI collagen, agrin, unknown endothelial antigens (Jackson et al. 2011b), and ICAM-1 (Lawson et al. 2005).
Alloantibodies mainly induce AMR by complement-dependent mechanisms. In fact, the alloantibodies that fix C1q complement are associated with a worse outcome that does not fix complement (Yabu et al. 2011). A greater risk of acute humoral rejection was clearly shown when C4d deposits were associated with DSA and, more specifically, anti-HLA class I but not anti-HLA class II antibodies (Cosio et al. 2010). However, in the absence of C4d staining, association of DSA with altered expression of endothelial genes was also found to be a marker of AMR in kidney patients, as the alloantibodies modified the microcirculation (Sis et al. 2009). NK cells and macrophages were also involved in AMR in patients with DSA. On stimulation by alloantibodies, NK cells cause endothelial injury and act as effector cells by killing target cells through the antibody-dependent cellular cytotoxicity pathway (Hidalgo et al. 2010). The role of NK cells is particularly well-defined as NKG2D is a receptor for stress-inducible MICA and, as described previously, this molecule is also highly efficient at stimulating NK cells and certain T-cell subsets (Bauer et al. 1999).
Several desensitizing protocols have been tested to reduce the DSA that cause AMR, including plasmapheresis or IVIg, as well as treatments targeting complement C5 molecules, proteasome, or CD20+ cells (Gueler et al. 2008; Raedler et al. 2011; Yoo et al. 2012). A combination of these treatments was shown to improve graft survival outcome (Lefaucheur et al. 2009; Montgomery et al. 2011).
CHRONIC REJECTION
The Origins and Mechanisms of Chronic Rejection
Since the early 1980s, 1-year graft survival of kidney allografts has increased significantly, reaching more than 90% (Hariharan et al. 2000; Pascual et al. 2002). Nevertheless, the graft survival in the long term has changed minimally, and the percentage of grafts lost annually after the first year has not changed (Meier-Kriesche et al. 2004). Death with a functioning graft and chronic allograft nephropathy (CAN) are the main causes of graft loss (Halloran et al. 1999; Ojo et al. 2000; Matas et al. 2002; Pascual et al. 2002). The predominance of CAN is 60%–70% in the first year posttransplant (Solez et al. 1998; Nankivell et al. 2003). CAN is an entity that encompasses several different mechanisms that include interstitial fibrosis and tubular atrophy. The natural history of CAN suggests that it can result from both immunological and nonimmunological phenomena, including calcineurin inhibitors (CNI) toxicity (Nankivell et al. 2003). Chronic dysfunction is also observed in other organ transplants and is often responsible for premature graft loss.
In recent years, significant efforts have been made to describe and classify CAN. In 1990, analyses based in histological studies were performed in the field of kidney transplantation, resulting in the description of a classification system for chronic allograft dysfunction (Classification “Banff 97”) (Racusen et al. 1999). This classification is revisited regularly to incorporate the latest scientific data and to try to differentiate lesions associated with chronic rejection (CR) from those observed in response to cardiovascular risk factors or CNI toxicity. The 2005 Banff meeting differentiated the lesions suggestive of chronic allograft rejection (including antibody-dependent complement activation lesions) and cell arteritis from the less specific lesions of interstitial fibrosis/tubular atrophy (IF/TA) (Solez et al. 2007, 2008). IF/TA lesions can appear very early after transplantation. At 1 year posttransplant, >80% of kidneys have minimal lesions of IF/TA that will deteriorate over time, reaching >50% of kidneys with severe lesions at 5 years. The 2011 Banff meeting focused on refining the diagnostic criteria for AMR. As such, C4d-negative AMR was recognized, in which NK cells and endothelial activation were proposed to play a role (Mengel et al. 2012).
The Histological Lesions
Significant progress has been made in understanding the mechanisms involved in the onset of CR, with the description of arteritis lesions in animal models (Yuan et al. 2002). One predominant observation made during chronic allograft rejection is an increase in the thickness of the intima, resulting in a decrease in vessel caliber with destruction of the internal elastic lamina. This thickening is also because of an accumulation of extracellular matrix and proliferation of myofibroblasts (Pedagogos et al. 1997; Pilmore et al. 2000; Ramirez et al. 2006). An accumulation of macrophages and CD4+ T cells has been observed at the periphery of the vessels (Thaunat and Nicoletti 2008; Thaunat et al. 2005, 2006, 2008), whereas CD8+ T cells are rarely present. The involvement of T cells in the pathogenesis of CR has been investigated through the study of genetically invalidated mice. In a mouse model of cardiac allotransplantation in which the genes encoding CD40 or CD40L were invalidated, CR was not observed, suggesting that activated T lymphocytes are required to initiate the phenomenon of CR. Nevertheless, in a rat model of cardiac allotransplantation in which animals were treated with CD40Ig (a molecule interfering with CD40-CD40L interaction), acute rejection was successfully inhibited, but CR occurred in long-term-surviving animals (Guillot et al. 2002). In this same model, the development of CR was prevented by simultaneous blockade of other costimulation pathways, such as RANK and ICOS (Guillonneau et al. 2004, 2005).
In some cases, the graft infiltrate has been found to adopt an organization resembling that of ectopic lymphoid tissues, with the infiltrating B cells capable of producing antidonor antibodies. These lesions of tertiary lymphoid tissue have been identified in kidney transplants, and the process is referred to as lymphoid neogenesis, which is analogous to the ontogenic program triggered during embryo development, the ectopic germinal centers (eGCs) (Thaunat et al. 2010; Cheng et al. 2011). The eGCs participate in the mechanisms of CR maintaining local memory T and B cells capable of synthesizing cytokines and antibodies. Interestingly, there is minimal overlap between the intragraft humoral response, suggesting a local antibody repertoire. Recently, Thaunat and coworkers showed that a breakdown of B-cell tolerance occurs within the graft during CR (Thaunat et al. 2012). It is thought that the micrograft environment interferes with peripheral deletion of autoreactive immature B cells that, in turn, produce autoantibodies. Recent studies have shown an important role for autoimmunity in the pathogenesis of CR suggesting a cross talk between the alloimmune response and autoimmunity (Sarma et al. 2012). Although all types of Th polarization profiles can lead to terminal CR, Thaunat and coworkers (Deteix et al. 2010) reported on a correlation between shorter graft survival and the presence of Th17 cells that produce IL-17 and IL-21 in human transplant patients. The latter group hypothesized that IL-21 promotes intragraft lymphoid neogenesis, which in turn supports the development of a local humoral immune response. Moreover, Burlingham et al. (Burlingham et al. 2007; Keller and Burlingham 2011) showed that the alloimmune response is partly responsible for the development of a Th17 autoimmune response. In this context, Fukami et al. (2009) showed that an anti-IL-17 therapy decreases the production of autoantibodies and the development of CR in a murine model.
The Role of Antibodies and Endothelial Cells
The involvement of antibodies directed against the graft in chronic rejection has been suggested by several groups showing a negative correlation between the appearance of antidonor antibodies and graft survival (Mao et al. 2007a,b; Sis et al. 2007). The emergence of new techniques to identify antibodies by fluorescence-activated cell sorting, flow cytometry, ELISA (enzyme-linked immunosorbent assay), and the most sensitive Luminex technique (flow cytometry with microbeads coated with peptides) now enables a better analysis of the humoral component (Pei et al. 2003). Moreover, recent data showing the existence of deposits during CR suggest the involvement of anti-HLA antibodies with the ability to activate complement (Nickeleit et al. 2002; Regele et al. 2002). However, observations from ABO-incompatible transplants, a context in which complement activation occurs, suggest the existence of adaptation mechanisms, allowing endothelial cells to resist complement activation (Gloor et al. 2006).
However, the effects of antibodies are diverse as described in Figure 2, and not limited to complement activation. Antibodies can also bind to the surface molecules of target cells and recruit other cells by interactions via their constant domain (Rebellato et al. 2006; Won et al. 2006). Recently, Hirohashi et al. described a new role for NK cells in chronic allograft rejection in which donor-specific antibodies interact with NK cells in the absence of complement and trigger transplant vasculopathy (Hirohashi et al. 2012; Li and Baldwin 2012). Last, there is some in vitro data showing that the culture of endothelial cells with antidonor antibodies results in the activation and proliferation of endothelial cells (Bian and Reed 1999).
Figure 2.
Induction of endothelial cells activation by antidonor antibodies. Antidonor antibodies (Abs) are known to induce chronic allograft rejection by several mechanisms of action involving their constant domain (Fc) or their capacity to induce activation of complement. Antidonor Abs also induce endothelial cell activation. Activated ECs secrete notably growth factors (GF) that induce the recruitment and activation/proliferation of several cells implicated in allograft rejection.
This activation step of endothelial cells is associated with the expression of various receptors on their cell surface (PDGF-R, EGF-R, FGF-R), as well as the synthesis of numerous growth factors (PDGF, EGF, FGF, VEGF, TGF-β, etc.) and synthesis of endothelin I (Bian and Reed 2001; Chen et al. 2001; Rossini et al. 2005). The local presence of growth factor is increased by the platelet adhesion that occurs during endothelial-cell activation, resulting in the generation of a local amplification loop with the release of many growth factors such as PDGF and TGF-β (MacDermott 1996; Yang et al. 2005). This phase of endothelial-cell activation promotes the stimulation of smooth muscle cells via release of endothelin I, and indirectly via the local synthesis of angiotensin II. It also causes local recruitment of inflammatory cells through the release of chemokines (MCP-1, IP-10), local activation of complement, local coagulation activation, promoting platelet adhesion, and releasing thromboxane A2. Finally, endothelial-cell activation stimulates the differentiation and proliferation of myofibroblast cells that synthesize the extracellular matrix involved in CR (Abbate et al. 2002; Cogan et al. 2002; Dewald et al. 2005; Li et al. 2007; Frangogiannis 2008; Haurani et al. 2008; Kennard et al. 2008; Wynn 2008). Myofibroblastic cells are important constituents of CR as they infiltrate the vessel wall and interstitial space. These cells express different cytoskeletal proteins (vimentin, α smooth muscle actin) as well as the myosin light chain, but do not express markers of lymphoid or epithelial cells (E-cadherin, ZO-1) (Badid et al. 2002). The origin of these cells is diverse. They originate from circulating stem cells capable of differentiating into endothelial cells or myocardiocytes (Direkze et al. 2003; Li et al. 2007). They can also originate from the transdifferentiation of endothelial cells or from renal tubular epithelial cells that transdifferentiate into myofibroblasts (Gressner 1996; Sommer et al. 2005; Hertig et al. 2006). The mechanisms involved remain poorly understood. However, the changes induced by ischemia/reperfusion or during CR appear to promote epithelial-mesenchymal transdifferentiation. TGF-β was also shown to be important for the promotion of epithelial-mesenchymal transdifferentiation (Fan et al. 1999; Mezzano et al. 2003; Lindert et al. 2005; Jiang et al. 2006; Meyer-ter-Vehn et al. 2006). Cell differentiation into myofibroblasts and expansion involves various growth factors including TGF-β, FGF, PDGF, IGF-1, angiotensin II, MCP-1, RANTES, TNF-α, IL-15, and connective tissue growth factor.
The Antigenic Targets of Antigens (Antidonor and Autoantibodies)
One of the independent risk factors for the development of chronic rejection is the presence of anti-HLA class I and especially class II antibodies (Ozawa et al. 2007). More than 80% of patients with transplant glomerulopathy have anti-HLA antibodies, 85% being directed against class I or class II antigens (Gloor et al. 2007; Sis et al. 2007; Issa et al. 2008). Less than half of biopsies (40%) display deposits of C4d, suggesting that mechanisms other than complement activation may be associated with CR (Solez et al. 2008). Apart from antibodies directed against the major histocompatibility complex HLA, other non-HLA antibodies may also contribute to the structural changes observed during chronic rejection. These include anti-MICA, anti-MICB (MHC class I-related molecules A and B), antiendothelial cells, antivimentin, and other autoantibodies directed against angiotensin II receptor (Dragun et al. 2005, 2008; Hilbrands et al. 2005; Zafar et al. 2006; Panigrahi et al. 2007; Zou et al. 2007; Baid-Agrawal and Frei 2008; Kamoun and Grossman 2008; Sumitran-Holgersson 2008).
Tissue Fibrosis
During the various immunological and nonimmunological rejection processes, several cell types, particularly endothelial cells, are responsible for the development of an extracellular matrix through the release of growth factors or cytokines such as endothelin I, angiotensin II, TNF, PDGF, TGF, etc. (Coupes et al. 1994; Lu et al. 2002; el-Agroudy et al. 2003; Baczkowska et al. 2005; Summers et al. 2005; Roos-van Groningen et al. 2006). Extracellular matrix accumulation is a lesion of fibrosis. In recent years, it has become apparent that tissue fibrosis is a dynamic equilibrium. Activation of different tissue protease, such as metalloproteinases, or the use of blocking molecule should prevent fibrosis and degrade extracellular matrix. These approaches must be validated in clinical models.
In conclusion, although significant progress has been made in developing immunosuppressive therapies that reduce the occurrence of acute rejection and improve allograft survival at 1 year, little progress has been made regarding graft survival in the longer term. Long-term survival is limited by complex and intricate mechanisms including chronic rejection, immunosuppressive drug toxicity, cardiovascular factors, dyslipidemia, diabetes, and so on. Understanding the mechanisms involved in the onset of chronic rejection should be promoted by approaches such as DNA chips or protein chips to better understand the different stages of development of chronic rejection and the different factors involved. This understanding should help develop new preventive and/or therapeutic approaches in organ transplantation.
Footnotes
Editors: Laurence A. Turka and Kathryn J. Wood
Additional Perspectives on Transplantation available at www.perspectivesinmedicine.org
REFERENCES
- Abbate M, Zoja C, Rottoli D, Corna D, Tomasoni S, Remuzzi G 2002. Proximal tubular cells promote fibrogenesis by TGF-β1-mediated induction of peritubular myofibroblasts. Kidney Int 61: 2066–2077 [DOI] [PubMed] [Google Scholar]
- Abdi R, Smith RN, Makhlouf L, Najafian N, Luster AD, Auchincloss H Jr, Sayegh MH 2002. The role of CC chemokine receptor 5 (CCR5) in islet allograft rejection. Diabetes 51: 2489–2495 [DOI] [PubMed] [Google Scholar]
- Abdi R, Means TK, Ito T, Smith RN, Najafian N, Jurewicz M, Tchipachvili V, Charo I, Auchincloss H Jr, Sayegh MH, et al. 2004. Differential role of CCR2 in islet and heart allograft rejection: Tissue specificity of chemokine/chemokine receptor function in vivo. J Immunol 172: 767–775 [DOI] [PubMed] [Google Scholar]
- Agostini C, Calabrese F, Rea F, Facco M, Tosoni A, Loy M, Binotto G, Valente M, Trentin L, Semenzato G 2001. Cxcr3 and its ligand CXCL10 are expressed by inflammatory cells infiltrating lung allografts and mediate chemotaxis of T cells at sites of rejection. Am J Pathol 158: 1703–1711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akashi S, Sho M, Kashizuka H, Hamada K, Ikeda N, Kuzumoto Y, Tsurui Y, Nomi T, Mizuno T, Kanehiro H, et al. 2005. A novel small-molecule compound targeting CCR5 and CXCR3 prevents acute and chronic allograft rejection. Transplantation 80: 378–384 [DOI] [PubMed] [Google Scholar]
- Akbar AN, Amlot PL, Timms A, Lombardi G, Lechler R, Janossy G 1990. The development of primed/memory CD8+ lymphocytes in vitro and in rejecting kidneys after transplantation. Clin Exp Immunol 81: 225–231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alferink J, Lieberam I, Reindl W, Behrens A, Weiss S, Huser N, Gerauer K, Ross R, Reske-Kunz AB, Ahmad-Nejad P, et al. 2003. Compartmentalized production of CCL17 in vivo: Strong inducibility in peripheral dendritic cells contrasts selective absence from the spleen. J Exp Med 197: 585–599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amano H, Bickerstaff A, Orosz CG, Novick AC, Toma H, Fairchild RL 2005. Absence of recipient CCR5 promotes early and increased allospecific antibody responses to cardiac allografts. J Immunol 174: 6499–6508 [DOI] [PubMed] [Google Scholar]
- Anegon I, Cuturi MC, Trinchieri G, Perussia B 1988. Interaction of Fc receptor (CD16) ligands induces transcription of interleukin 2 receptor (CD25) and lymphokine genes and expression of their products in human natural killer cells. J Exp Med 167: 452–472 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anglicheau D, Sharma VK, Ding R, Hummel A, Snopkowski C, Dadhania D, Seshan SV, Suthanthiran M 2009. MicroRNA expression profiles predictive of human renal allograft status. Proc Natl Acad Sci 106: 5330–5335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aquino-Dias EC, Joelsons G, da Silva DM, Berdichevski RH, Ribeiro AR, Veronese FJ, Goncalves LF, Manfro RC 2008. Non-invasive diagnosis of acute rejection in kidney transplants with delayed graft function. Kidney Int 73: 877–884 [DOI] [PubMed] [Google Scholar]
- Ashton-Chess J, Giral M, Mengel M, Renaudin K, Foucher Y, Gwinner W, Braud C, Dugast E, Quillard T, Thebault P, et al. 2008. Tribbles-1 as a novel biomarker of chronic antibody-mediated rejection. J Am Soc Nephrol 19: 1116–1127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashton-Chess J, Dugast E, Colvin RB, Giral M, Foucher Y, Moreau A, Renaudin K, Braud C, Devys A, Brouard S, et al. 2009. Regulatory, effector, and cytotoxic T cell profiles in long-term kidney transplant patients. J Am Soc Nephrol 20: 1113–1122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashton-Chess J, Mai HL, Jovanovic V, Renaudin K, Foucher Y, Giral M, Moreau A, Dugast E, Mengel M, Racape M, et al. 2010. Immunoproteasome β subunit 10 is increased in chronic antibody-mediated rejection. Kidney Int 77: 880–890 [DOI] [PubMed] [Google Scholar]
- Ayalon O, Hughes EA, Cresswell P, Lee J, O’Donnell L, Pardi R, Bender JR 1998. Induction of transporter associated with antigen processing by interferon γ confers endothelial cell cytoprotection against natural killer-mediated lysis. Proc Natl Acad Sci 95: 2435–2440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baczkowska T, Perkowska-Ptasinska A, Sadowska A, Lewandowski Z, Nowacka-Cieciura E, Cieciura T, Pazik J, Lewandowska D, Mroz A, Urbanowicz A, et al. 2005. Serum TGF-β1 correlates with chronic histopathological lesions in protocol biopsies of kidney allograft recipients. Transplant Proc 37: 773–775 [DOI] [PubMed] [Google Scholar]
- Badid C, Desmouliere A, Babici D, Hadj-Aissa A, McGregor B, Lefrancois N, Touraine JL, Laville M 2002. Interstitial expression of α-SMA: An early marker of chronic renal allograft dysfunction. Nephrol Dial Transplant 17: 1993–1998 [DOI] [PubMed] [Google Scholar]
- Baid-Agrawal S, Frei UA 2008. Kidney-transplant rejection and anti-MICA antibodies. N Engl J Med 358: 196. [PubMed] [Google Scholar]
- Barbara JA, Turvey SE, Kingsley CI, Spriewald BM, Hara M, Witzke O, Morris PJ, Wood KJ 2000. Islet allograft rejection can be mediated by CD4+, alloantigen experienced, direct pathway T cells of TH1 and TH2 cytokine phenotype. Transplantation 70: 1641–1649 [DOI] [PubMed] [Google Scholar]
- Barber DL, Wherry EJ, Ahmed R 2003. Cutting edge: Rapid in vivo killing by memory CD8 T cells. J Immunol 171: 27–31 [DOI] [PubMed] [Google Scholar]
- Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T 1999. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285: 727–729 [DOI] [PubMed] [Google Scholar]
- Belperio JA, Ardehali A 2008. Chemokines and transplant vasculopathy. Circ Res 103: 454–466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bestard O, Cruzado JM, Mestre M, Caldes A, Bas J, Carrera M, Torras J, Rama I, Moreso F, Seron D, et al. 2007. Achieving donor-specific hyporesponsiveness is associated with FoxP3+ regulatory T cell recruitment in human renal allograft infiltrates. J Immunol 179: 4901–4909 [DOI] [PubMed] [Google Scholar]
- Bian H, Reed EF 1999. Alloantibody-mediated class I signal transduction in endothelial cells and smooth muscle cells: Enhancement by IFN-γ and TNF-α. J Immunol 163: 1010–1018 [PubMed] [Google Scholar]
- Bian H, Reed EF 2001. Anti-HLA class I antibodies transduce signals in endothelial cells resulting in FGF receptor translocation, down-regulation of ICAM-1 and cell proliferation. Transplant Proc 33: 311. [DOI] [PubMed] [Google Scholar]
- Brandle D, Joergensen J, Zenke G, Burki K, Hof RP 1998. Contribution of donor-specific antibodies to acute allograft rejection: Evidence from B cell-deficient mice. Transplantation 65: 1489–1493 [DOI] [PubMed] [Google Scholar]
- Braun MY, Desalle F, Le Moine A, Pretolani M, Matthys P, Kiss R, Goldman M 2000. IL-5 and eosinophils mediate the rejection of fully histoincompatible vascularized cardiac allografts: Regulatory role of alloreactive CD8+ T lymphocytes and IFN-γ. Eur J Immunol 30: 1290–1296 [DOI] [PubMed] [Google Scholar]
- Brook MO, Wood JK, Jones ND 2006. The impact of memory T cells on rejection and the induction of tolerance. Transplantation 82: 1–9 [DOI] [PubMed] [Google Scholar]
- Burlingham WJ, Love RB, Jankowska-Gan E, Haynes LD, Xu Q, Bobadilla JL, Meyer KC, Hayney MS, Braun RK, Greenspan DS, et al. 2007. IL-17-dependent cellular immunity to collagen type V predisposes to obliterative bronchiolitis in human lung transplants. J Clin Invest 117: 3498–3506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell JJ, Haraldsen G, Pan J, Rottman J, Qin S, Ponath P, Andrew DP, Warnke R, Ruffing N, Kassam N, et al. 1999. The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T cells. Nature 400: 776–780 [DOI] [PubMed] [Google Scholar]
- Castellano G, Melchiorre R, Loverre A, Ditonno P, Montinaro V, Rossini M, Divella C, Battaglia M, Lucarelli G, Annunziata GS, et al. 2010. Therapeutic targeting of classical and lectin pathways of complement protects from ischemia-reperfusion-induced renal damage. Am J Pathol 176: 1648–1659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celli S, Albert ML, Bousso P 2011. Visualizing the innate and adaptive immune responses underlying allograft rejection by two-photon microscopy. Nat Med 17: 744–749 [DOI] [PubMed] [Google Scholar]
- Chen J, Fabry B, Schiffrin EL, Wang N 2001. Twisting integrin receptors increases endothelin-1 gene expression in endothelial cells. Am J Physiol Cell Physiol 280: C1475–C1484 [DOI] [PubMed] [Google Scholar]
- Cheng J, Torkamani A, Grover RK, Jones TM, Ruiz DI, Schork NJ, Quigley MM, Hall FW, Salomon DR, Lerner RA 2011. Ectopic B-cell clusters that infiltrate transplanted human kidneys are clonal. Proc Natl Acad Sci 108: 5560–5565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chong AS, Alegre ML 2012. The impact of infection and tissue damage in solid-organ transplantation. Nat Rev Immunol 12: 459–471 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christopher K, Mueller TF, Ma C, Liang Y, Perkins DL 2002. Analysis of the innate and adaptive phases of allograft rejection by cluster analysis of transcriptional profiles. J Immunol 169: 522–530 [DOI] [PubMed] [Google Scholar]
- Clatworthy MR, Watson CJ, Plotnek G, Bardsley V, Chaudhry AN, Bradley JA, Smith KG 2009. B-cell-depleting induction therapy and acute cellular rejection. N Engl J Med 360: 2683–2685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cogan JG, Subramanian SV, Polikandriotis JA, Kelm RJ Jr, Strauch AR 2002. Vascular smooth muscle α-actin gene transcription during myofibroblast differentiation requires Sp1/3 protein binding proximal to the MCAT enhancer. J Biol Chem 277: 36433–36442 [DOI] [PubMed] [Google Scholar]
- Colobran R, Casamitjana N, Roman A, Faner R, Pedrosa E, Arostegui JI, Pujol-Borrell R, Juan M, Palou E 2009. Copy number variation in the CCL4L gene is associated with susceptibility to acute rejection in lung transplantation. Genes Immun 10: 254–259 [DOI] [PubMed] [Google Scholar]
- Constant SL 1999. B lymphocytes as antigen-presenting cells for CD4+ T cell priming in vivo. J Immunol 162: 5695–5703 [PubMed] [Google Scholar]
- Cooper JE, Gralla J, Cagle L, Goldberg R, Chan L, Wiseman AC 2011. Inferior kidney allograft outcomes in patients with de novo donor-specific antibodies are due to acute rejection episodes. Transplantation 91: 1103–1109 [DOI] [PubMed] [Google Scholar]
- Cosio FG, Lager DJ, Lorenz EC, Amer H, Gloor JM, Stegall MD 2010. Significance and implications of capillaritis during acute rejection of kidney allografts. Transplantation 89: 1088–1094 [DOI] [PubMed] [Google Scholar]
- Coudert JD, Coureau C, Guery JC 2002. Preventing NK cell activation by donor dendritic cells enhances allospecific CD4 T cell priming and promotes Th type 2 responses to transplantation antigens. J Immunol 169: 2979–2987 [DOI] [PubMed] [Google Scholar]
- Coupel S, Moreau A, Hamidou M, Horejsi V, Soulillou JP, Charreau B 2007. Expression and release of soluble HLA-E is an immunoregulatory feature of endothelial cell activation. Blood 109: 2806–2814 [DOI] [PubMed] [Google Scholar]
- Coupes BM, Newstead CG, Short CD, Brenchley PE 1994. Transforming growth factor β1 in renal allograft recipients. Transplantation 57: 1727–1731 [PubMed] [Google Scholar]
- Crescioli C, Buonamano A, Scolletta S, Sottili M, Francalanci M, Giomarelli P, Biagioli B, Lisi G, Pradella F, Serio M, et al. 2009. Predictive role of pretransplant serum CXCL10 for cardiac acute rejection. Transplantation 87: 249–255 [DOI] [PubMed] [Google Scholar]
- Croft M 2003. Costimulation of T cells by OX40, 4-1BB, and CD27. Cytokine Growth Factor Rev 14: 265–273 [DOI] [PubMed] [Google Scholar]
- Cuturi MC, Murphy M, Costa-Giomi MP, Weinmann R, Perussia B, Trinchieri G 1987. Independent regulation of tumor necrosis factor and lymphotoxin production by human peripheral blood lymphocytes. J Exp Med 165: 1581–1594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cuturi MC, Anegon I, Sherman F, Loudon R, Clark SC, Perussia B, Trinchieri G 1989. Production of hematopoietic colony-stimulating factors by human natural killer cells. J Exp Med 169: 569–583 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deng S, Moore Dj, Huang X, Lian MM, Mohiuddin M, Velededeoglu E, Lee MKT, Sonawane S, Kim J, Wang J, et al. 2007. Cutting edge: Transplant tolerance induced by anti-CD45RB requires B lymphocytes. J Immunol 178: 6028–6032 [DOI] [PubMed] [Google Scholar]
- Deteix C, Attuil-Audenis V, Duthey A, Patey N, McGregor B, Dubois V, Caligiuri G, Graff-Dubois S, Morelon E, Thaunat O 2010. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J Immunol 184: 5344–5351 [DOI] [PubMed] [Google Scholar]
- Dewald O, Zymek P, Winkelmann K, Koerting A, Ren G, Abou-Khamis T, Michael LH, Rollins BJ, Entman ML, Frangogiannis NG 2005. CCL2/monocyte chemoattractant protein-1 regulates inflammatory responses critical to healing myocardial infarcts. Circ Res 96: 881–889 [DOI] [PubMed] [Google Scholar]
- Dikow R, Becker LE, Schaier M, Waldherr R, Gross ML, Zeier M 2010. In renal transplants with delayed graft function chemokines and chemokine receptor expression predict long-term allograft function. Transplantation 90: 771–776 [DOI] [PubMed] [Google Scholar]
- Dilek N, Poirier N, Usal C, Martinet B, Blancho G, Vanhove B 2012. Control of transplant tolerance and intragraft regulatory T cell localization by myeloid-derived suppressor cells and CCL5. J Immunol 188: 4209–4216 [DOI] [PubMed] [Google Scholar]
- DiLillo DJ, Griffiths R, Seshan SV, Magro CM, Ruiz P, Coffman TM, Tedder TF 2011. B lymphocytes differentially influence acute and chronic allograft rejection in mice. J Immunol 186: 2643–2654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding R, Li B, Muthukumar T, Dadhania D, Medeiros M, Hartono C, Serur D, Seshan SV, Sharma VK, Kapur S, et al. 2003. CD103 mRNA levels in urinary cells predict acute rejection of renal allografts. Transplantation 75: 1307–1312 [DOI] [PubMed] [Google Scholar]
- Ding Q, Yeung M, Camirand G, Zeng Q, Akiba H, Yagita H, Chalasani G, Sayegh MH, Najafian N, Rothstein DM 2011. Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice. J Clin Invest 121: 3645–3656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Direkze NC, Forbes SJ, Brittan M, Hunt T, Jeffery R, Preston SL, Poulsom R, Hodivala-Dilke K, Alison MR, Wright NA 2003. Multiple organ engraftment by bone-marrow-derived myofibroblasts and fibroblasts in bone-marrow-transplanted mice. Stem Cells 21: 514–520 [DOI] [PubMed] [Google Scholar]
- Dragun D 2008. Humoral responses directed against non-human leukocyte antigens in solid-organ transplantation. Transplantation 86: 1019–1025 [DOI] [PubMed] [Google Scholar]
- Dragun D, Muller DN, Brasen JH, Fritsche L, Nieminen-Kelha M, Dechend R, Kintscher U, Rudolph B, Hoebeke J, Eckert D, et al. 2005. Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection. N Engl J Med 352: 558–569 [DOI] [PubMed] [Google Scholar]
- Dragun D, Scornik J, Meier-Kriesche HU 2008. Kidney-transplant rejection and anti-MICA antibodies. N Engl J Med 358: 196; author reply 196 [PubMed] [Google Scholar]
- Dugre FJ, Gaudreau S, Belles-Isles M, Houde I, Roy R 2000. Cytokine and cytotoxic molecule gene expression determined in peripheral blood mononuclear cells in the diagnosis of acute renal rejection. Transplantation 70: 1074–1080 [DOI] [PubMed] [Google Scholar]
- Duluc D, Delneste Y, Tan F, Moles MP, Grimaud L, Lenoir J, Preisser L, Anegon I, Catala L, Ifrah N, et al. 2007. Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 110: 4319–4330 [DOI] [PubMed] [Google Scholar]
- el-Agroudy AE, Hassan NA, Foda MA, Ismail AM, el-Sawy EA, Mousa O, Ghoneim MA 2003. Effect of angiotensin II receptor blocker on plasma levels of TGF-β1 and interstitial fibrosis in hypertensive kidney transplant patients. Am J Nephrol 23: 300–306 [DOI] [PubMed] [Google Scholar]
- El-Sawy T, Belperio JA, Strieter RM, Remick DG, Fairchild RL 2005. Inhibition of polymorphonuclear leukocyte-mediated graft damage synergizes with short-term costimulatory blockade to prevent cardiac allograft rejection. Circulation 112: 320–331 [DOI] [PubMed] [Google Scholar]
- Epperson DE, Pober JS 1994. Antigen-presenting function of human endothelial cells. Direct activation of resting CD8 T cells. J Immunol 153: 5402–5412 [PubMed] [Google Scholar]
- Fahim T, Bohmig GA, Exner M, Huttary N, Kerschner H, Kandutsch S, Kerjaschki D, Brambock A, Nagy-Bojarszky K, Regele H 2007. The cellular lesion of humoral rejection: Predominant recruitment of monocytes to peritubular and glomerular capillaries. Am J Transplant 7: 385–393 [DOI] [PubMed] [Google Scholar]
- Fahmy NM, Yamani MH, Starling RC, Ratliff NB, Young JB, McCarthy PM, Feng J, Novick AC, Fairchild RL 2003. Chemokine and receptor-gene expression during early and late acute rejection episodes in human cardiac allografts. Transplantation 75: 2044–2047 [DOI] [PubMed] [Google Scholar]
- Fan JM, Ng YY, Hill PA, Nikolic-Paterson DJ, Mu W, Atkins RC, Lan HY 1999. Transforming growth factor-β regulates tubular epithelial-myofibroblast transdifferentiation in vitro. Kidney Int 56: 1455–1467 [DOI] [PubMed] [Google Scholar]
- Farid WR, Pan Q, van der Meer AJ, de Ruiter PE, Ramakrishnaiah V, de Jonge J, Kwekkeboom J, Janssen HL, Metselaar HJ, Tilanus HW, et al. 2012. Hepatocyte-derived microRNAs as serum biomarkers of hepatic injury and rejection after liver transplantation. Liver Transpl 18: 290–297 [DOI] [PubMed] [Google Scholar]
- Fong AM, Robinson LA, Steeber DA, Tedder TF, Yoshie O, Imai T, Patel DD 1998. Fractalkine and CX3CR1 mediate a novel mechanism of leukocyte capture, firm adhesion, and activation under physiologic flow. J Exp Med 188: 1413–1419 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foussat A, Coulomb-L’Hermine A, Gosling J, Krzysiek R, Durand-Gasselin I, Schall T, Balian A, Richard Y, Galanaud P, Emilie D 2000. Fractalkine receptor expression by T lymphocyte subpopulations and in vivo production of fractalkine in human. Eur J Immunol 30: 87–97 [DOI] [PubMed] [Google Scholar]
- Frangogiannis NG 2008. The immune system and cardiac repair. Pharmacol Res 58: 88–111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman BH, Wolf JH, Wang L, Putt ME, Shaked A, Christie JD, Hancock WW, Olthoff KM 2012. Serum cytokine profiles associated with early allograft dysfunction in patients undergoing liver transplantation. Liver Transpl 18: 166–176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fukami N, Ramachandran S, Saini D, Walter M, Chapman W, Patterson GA, Mohanakumar T 2009. Antibodies to MHC class I induce autoimmunity: Role in the pathogenesis of chronic rejection. J Immunol 182: 309–318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao W, Faia KL, Csizmadia V, Smiley ST, Soler D, King JA, Danoff TM, Hancock WW 2001. Beneficial effects of targeting CCR5 in allograft recipients. Transplantation 72: 1199–1205 [DOI] [PubMed] [Google Scholar]
- Garrod KR, Liu FC, Forrest LE, Parker I, Kang SM, Cahalan MD 2010. NK cell patrolling and elimination of donor-derived dendritic cells favor indirect alloreactivity. J Immunol 184: 2329–2336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gelman AE, Okazaki M, Sugimoto S, Li W, Kornfeld CG, Lai J, Richardson SB, Kreisel FH, Huang HJ, Tietjens JR, et al. 2010. CCR2 regulates monocyte recruitment as well as CD4 T1 allorecognition after lung transplantation. Am J Transplant 10: 1189–1199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gloor JM, Cosio FG, Rea DJ, Wadei HM, Winters JL, Moore SB, DeGoey SR, Lager DJ, Grande JP, Stegall MD 2006. Histologic findings one year after positive crossmatch or ABO blood group incompatible living donor kidney transplantation. Am J Transplant 6: 1841–1847 [DOI] [PubMed] [Google Scholar]
- Gloor JM, Sethi S, Stegall MD, Park WD, Moore SB, DeGoey S, Griffin MD, Larson TS, Cosio FG 2007. Transplant glomerulopathy: Subclinical incidence and association with alloantibody. Am J Transplant 7: 2124–2132 [DOI] [PubMed] [Google Scholar]
- Gregson AL, Hoji A, Saggar R, Ross DJ, Kubak BM, Jamieson BD, Weigt SS, Lynch JP III, Ardehali A, Belperio JA, et al. 2008. Bronchoalveolar immunologic profile of acute human lung transplant allograft rejection. Transplantation 85: 1056–1059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gressner AM 1996. Transdifferentiation of hepatic stellate cells (Ito cells) to myofibroblasts: A key event in hepatic fibrogenesis. Kidney Int Suppl 54: S39–S45 [PubMed] [Google Scholar]
- Griffin GK, Newton G, Tarrio ML, Bu DX, Maganto-Garcia E, Azcutia V, Alcaide P, Grabie N, Luscinskas FW, Croce KJ, et al. 2012. IL-17 and TNF-α sustain neutrophil recruitment during inflammation through synergistic effects on endothelial activation. J Immunol 188: 6287–6299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gueler F, Rong S, Gwinner W, Mengel M, Brocker V, Schon S, Greten TF, Hawlisch H, Polakowski T, Schnatbaum K, et al. 2008. Complement 5a receptor inhibition improves renal allograft survival. J Am Soc Nephrol 19: 2302–2312 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guillonneau C, Louvet C, Renaudin K, Heslan JM, Heslan M, Tesson L, Vignes C, Guillot C, Choi Y, Turka LA, et al. 2004. The role of TNF-related activation-induced cytokine-receptor activating NF-κB interaction in acute allograft rejection and CD40L-independent chronic allograft rejection. J Immunol 172:1619–1629 [DOI] [PubMed] [Google Scholar]
- Guillonneau C, Aubry V, Renaudin K, Seveno C, Usal C, Tezuka K, Anegon I 2005. Inhibition of chronic rejection and development of tolerogenic T cells after ICOS-ICOSL and CD40-CD40L co-stimulation blockade. Transplantation 80: 546–554 [PubMed] [Google Scholar]
- Guillonneau C, Hill M, Hubert FX, Chiffoleau E, Herve C, Li XL, Heslan M, Usal C, Tesson L, Menoret S, et al. 2007. CD40Ig treatment results in allograft acceptance mediated by CD8CD45RC T cells, IFN-γ, and indoleamine 2,3-dioxygenase. J Clin Invest 117: 1096–1106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guillonneau C, Picarda E, Anegon I 2010. CD8+ regulatory T cells in solid organ transplantation. Curr Opin Organ Transplant (to be published) [DOI] [PubMed] [Google Scholar]
- Guillot C, Guillonneau C, Mathieu P, Gerdes CA, Menoret S, Braudeau C, Tesson L, Renaudin K, Castro MG, Lowenstein PR, et al. 2002. Prolonged blockade of CD40-CD40 ligand interactions by gene transfer of CD40Ig results in long-term heart allograft survival and donor-specific hyporesponsiveness, but does not prevent chronic rejection. J Immunol 168: 1600–1609 [DOI] [PubMed] [Google Scholar]
- Halloran PF, Wadgymar A, Ritchie S, Falk J, Solez K, Srinivasa NS 1990. The significance of the anti-class I antibody response. I: Clinical and pathologic features of anti-class I-mediated rejection. Transplantation 49: 85–91 [DOI] [PubMed] [Google Scholar]
- Halloran PF, Melk A, Barth C 1999. Rethinking chronic allograft nephropathy: The concept of accelerated senescence. J Am Soc Nephrol 10: 167–181 [DOI] [PubMed] [Google Scholar]
- Hancock WW, Thomson NM, Atkins RC 1983. Composition of interstitial cellular infiltrate identified by monoclonal antibodies in renal biopsies of rejecting human renal allografts. Transplantation 35: 458–463 [DOI] [PubMed] [Google Scholar]
- Hancock WW, Lu B, Gao W, Csizmadia V, Faia K, King JA, Smiley ST, Ling M, Gerard NP, Gerard C 2000. Requirement of the chemokine receptor CXCR3 for acute allograft rejection. J Exp Med 192: 1515–1520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hariharan S, Johnson CP, Bresnahan BA, Taranto SE, McIntosh MJ, Stablein D 2000. Improved graft survival after renal transplantation in the United States, 1988 to 1996. N Engl J Med 342: 605–612 [DOI] [PubMed] [Google Scholar]
- Haurani MJ, Cifuentes ME, Shepard AD, Pagano PJ 2008. Nox4 oxidase overexpression specifically decreases endogenous Nox4 mRNA and inhibits angiotensin II-induced adventitial myofibroblast migration. Hypertension 52: 143–149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- He H, Stone JR, Perkins DL 2002. Analysis of robust innate immune response after transplantation in the absence of adaptive immunity. Transplantation 73: 853–861 [DOI] [PubMed] [Google Scholar]
- He H, Stone JR, Perkins DL, Perkins 2003. Analysis of differential immune responses induced by innate and adaptive immunity following transplantation. Immunology 109: 185–196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heeger PS, Greenspan NS, Kuhlenschmidt S, Dejelo C, Hricik DE, Schulak JA, Tary-Lehmann M 1999. Pretransplant frequency of donor-specific, IFN-γ-producing lymphocytes is a manifestation of immunologic memory and correlates with the risk of posttransplant rejection episodes. J Immunol 163: 2267–2275 [PubMed] [Google Scholar]
- Hengel RL, Thaker V, Pavlick MV, Metcalf JA, Dennis G Jr, Yang J, Lempicki RA, Sereti I, Lane HC 2003. Cutting edge: L-selectin (CD62L) expression distinguishes small resting memory CD4+ T cells that preferentially respond to recall antigen. J Immunol 170: 28–32 [DOI] [PubMed] [Google Scholar]
- Herrera OB, Golshayan D, Tibbott R, Salcido Ochoa F, James MJ, Marelli-Berg FM, Lechler RI 2004. A novel pathway of alloantigen presentation by dendritic cells. J Immunol 173: 4828–4837 [DOI] [PubMed] [Google Scholar]
- Hertig A, Verine J, Mougenot B, Jouanneau C, Ouali N, Sebe P, Glotz D, Ancel PY, Rondeau E, Xu-Dubois YC 2006. Risk factors for early epithelial to mesenchymal transition in renal grafts. Am J Transplant 6: 2937–2946 [DOI] [PubMed] [Google Scholar]
- Hidalgo LG, Sis B, Sellares J, Campbell PM, Mengel M, Einecke G, Chang J, Halloran PF 2010. NK cell transcripts and NK cells in kidney biopsies from patients with donor-specific antibodies: Evidence for NK cell involvement in antibody-mediated rejection. Am J Transplant 10: 1812–1822 [DOI] [PubMed] [Google Scholar]
- Hilbrands L, Hoitsma A, Wetzels J 2005. Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection. N Engl J Med 352: 2027–2028 [DOI] [PubMed] [Google Scholar]
- Hirohashi T, Chase CM, Della Pelle P, Sebastian D, Alessandrini A, Madsen JC, Russell PS, Colvin RB 2012. A novel pathway of chronic allograft rejection mediated by NK cells and alloantibody. Am J Transplant 12: 313–321 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ho J, Rush DN, Gibson IW, Karpinski M, Storsley L, Bestland J, Stefura W, HayGlass KT, Nickerson PW 2010. Early urinary CCL2 is associated with the later development of interstitial fibrosis and tubular atrophy in renal allografts. Transplantation 90: 394–400 [DOI] [PubMed] [Google Scholar]
- Hoerning A, Koss K, Datta D, Boneschansker L, Jones CN, Wong IY, Irimia D, Calzadilla K, Benitez F, Hoyer PF, et al. 2011. Subsets of human CD4+ regulatory T cells express the peripheral homing receptor CXCR3. Eur J Immunol 41: 2291–2302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffmann U, Segerer S, Rummele P, Kruger B, Pietrzyk M, Hofstadter F, Banas B, Kramer BK 2006. Expression of the chemokine receptor CXCR3 in human renal allografts—A prospective study. Nephrol Dial Transplant 21: 1373–1381 [DOI] [PubMed] [Google Scholar]
- Hoffmann U, Bergler T, Segerer S, Rummele P, Kruger B, Banas MC, Reinhold S, Banas B, Kramer BK 2010. Impact of chemokine receptor CX3CR1 in human renal allograft rejection. Transpl Immunol 23: 204–208 [DOI] [PubMed] [Google Scholar]
- Hopken UE, Droese J, Li JP, Joergensen J, Breitfeld D, Zerwes HG, Lipp M 2004. The chemokine receptor CCR7 controls lymph node-dependent cytotoxic T cell priming in alloimmune responses. Eur J Immunol 34: 461–470 [DOI] [PubMed] [Google Scholar]
- Horne PH, Zimmerer JM, Fisher MG, Lunsford KE, Nadasdy G, Nadasdy T, van Rooijen N, Bumgardner GL 2008. Critical role of effector macrophages in mediating CD4-dependent alloimmune injury of transplanted liver parenchymal cells. J Immunol 181: 1224–1231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu H, Kwun J, Aizenstein BD, Knechtle SJ 2009. Noninvasive detection of acute and chronic injuries in human renal transplant by elevation of multiple cytokines/chemokines in urine. Transplantation 87: 1814–1820 [DOI] [PubMed] [Google Scholar]
- Huser N, Tertilt C, Gerauer K, Maier S, Traeger T, Assfalg V, Reiter R, Heidecke CD, Pfeffer K 2005. CCR4-deficient mice show prolonged graft survival in a chronic cardiac transplant rejection model. Eur J Immunol 35: 128–138 [DOI] [PubMed] [Google Scholar]
- Imai T, Hieshima K, Haskell C, Baba M, Nagira M, Nishimura M, Kakizaki M, Takagi S, Nomiyama H, Schall TJ, et al. 1997. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell 91: 521–530 [DOI] [PubMed] [Google Scholar]
- Issa N, Cosio FG, Gloor JM, Sethi S, Dean PG, Moore SB, DeGoey S, Stegall MD 2008. Transplant glomerulopathy: Risk and prognosis related to anti-human leukocyte antigen class II antibody levels. Transplantation 86: 681–685 [DOI] [PubMed] [Google Scholar]
- Ito A, Shimura H, Nitahara A, Tomiyama K, Ito M, Kanekura T, Okumura K, Yagita H, Kawai K 2008. NK cells contribute to the skin graft rejection promoted by CD4+ T cells activated through the indirect allorecognition pathway. Int Immunol 20: 1343–1349 [DOI] [PubMed] [Google Scholar]
- Iwasaki A, Medzhitov R 2010. Regulation of adaptive immunity by the innate immune system. Science 327: 291–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson JA, Kim EJ, Begley B, Cheeseman J, Harden T, Perez SD, Thomas S, Warshaw B, Kirk AD 2011a. Urinary chemokines CXCL9 and CXCL10 are noninvasive markers of renal allograft rejection and BK viral infection. Am J Transplant 11: 2228–2234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson AM, Lucas DP, Melancon JK, Desai NM 2011b. Clinical relevance and IgG subclass determination of non-HLA antibodies identified using endothelial cell precursors isolated from donor blood. Transplantation 92: 54–60 [DOI] [PubMed] [Google Scholar]
- Ji J, Yu YY, Chen H, Deng XX, Shen BY, Zhou GW, Peng CW, Li HW 2004. Perforin and granzyme B expression in liver allograft rejection. Zhonghua Gan Zang Bing Za Zhi 12: 325–327 [PubMed] [Google Scholar]
- Jiang YL, Dai AG, Li QF, Hu RC 2006. Transforming growth factor-β1 induces transdifferentiation of fibroblasts into myofibroblasts in hypoxic pulmonary vascular remodeling. Acta Biochim Biophys Sin (Shanghai) 38: 29–36 [DOI] [PubMed] [Google Scholar]
- Jose MD, Le Meur Y, Atkins RC, Chadban SJ 2003. Blockade of macrophage colony-stimulating factor reduces macrophage proliferation and accumulation in renal allograft rejection. Am J Transplant 3: 294–300 [DOI] [PubMed] [Google Scholar]
- Jungraithmayr W, Inci I, Bain M, Hillinger S, Korom S, Weder W 2010. Distribution of macrophages and T cells in syngrafts and allografts after experimental rat lung transplantation. Immunobiology 215: 206–214 [DOI] [PubMed] [Google Scholar]
- Kakuta Y, Okumi M, Miyagawa S, Tsutahara K, Abe T, Yazawa K, Matsunami K, Otsuka H, Takahara S, Nonomura N 2012. Blocking of CCR5 and CXCR3 suppresses the infiltration of macrophages in acute renal allograft rejection. Transplantation 93: 24–31 [DOI] [PubMed] [Google Scholar]
- Kamoun M, Grossman RA 2008. Kidney-transplant rejection and anti-MICA antibodies. N Engl J Med 358: 195. [DOI] [PubMed] [Google Scholar]
- Kedainis RL, Koch MJ, Brennan DC, Liapis H 2009. Focal C4d+ in renal allografts is associated with the presence of donor-specific antibodies and decreased allograft survival. Am J Transplant 9: 812–819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keller MR, Burlingham WJ 2011. Loss of tolerance to self after transplant. Semin Immunopathol 33: 105–110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kennard S, Liu H, Lilly B 2008. Transforming growth factor-β (TGF-1) down-regulates Notch3 in fibroblasts to promote smooth muscle gene expression. J Biol Chem 283: 1324–1333 [DOI] [PubMed] [Google Scholar]
- Kim J, Chang CK, Hayden T, Liu FC, Benjamin J, Hamerman JA, Lanier LL, Kang SM 2007. The activating immunoreceptor NKG2D and its ligands are involved in allograft transplant rejection. J Immunol 179: 6416–6420 [DOI] [PubMed] [Google Scholar]
- Kingsley CI, Karim M, Bushell AR, Wood KJ 2002. CD25+ CD4+ regulatory T cells prevent graft rejection: CTLA-4- and IL-10-dependent immunoregulation of alloresponses. J Immunol 168: 1080–1086 [DOI] [PubMed] [Google Scholar]
- Kirk AD, Morrell CN, Baldwin WM III 2009. Platelets influence vascularized organ transplants from start to finish. Am J Transplant 9: 14–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kozakowski N, Bohmig GA, Exner M, Soleiman A, Huttary N, Nagy-Bojarszky K, Ecker RC, Kikic Z, Regele H 2009. Monocytes/macrophages in kidney allograft intimal arteritis: No association with markers of humoral rejection or with inferior outcome. Nephrol Dial Transplant 24: 1979–1986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krams SM, Villanueva JC, Quinn MB, Martinez OM 1995. Expression of the cytotoxic T cell mediator granzyme B during liver allograft rejection. Transpl Immunol 3: 162–166 [DOI] [PubMed] [Google Scholar]
- Kroemer A, Xiao X, Degauque N, Edtinger K, Wei H, Demirci G, Li XC 2008. The innate NK cells, allograft rejection, and a key role for IL-15. J Immunol 180: 7818–7826 [DOI] [PubMed] [Google Scholar]
- Kwun J, Hazinedaroglu SM, Schadde E, Kayaoglu HA, Fechner J, Hu HZ, Roenneburg D, Torrealba J, Shiao L, Hong X, et al. 2008. Unaltered graft survival and intragraft lymphocytes infiltration in the cardiac allograft of CXCR3−/− mouse recipients. Am J Transplant 8: 1593–1603 [DOI] [PubMed] [Google Scholar]
- Laffont S, Seillet C, Ortaldo J, Coudert JD, Guery JC 2008. Natural killer cells recruited into lymph nodes inhibit alloreactive T-cell activation through perforin-mediated killing of donor allogeneic dendritic cells. Blood 112: 661–671 [DOI] [PubMed] [Google Scholar]
- Land WG 2005. The role of postischemic reperfusion injury and other nonantigen-dependent inflammatory pathways in transplantation. Transplantation 79: 505–514 [DOI] [PubMed] [Google Scholar]
- LaRosa DF, Rahman AH, Turka LA 2007. The innate immune system in allograft rejection and tolerance. J Immunol 178:7503–7509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lawson C, Holder AL, Stanford RE, Smith J, Rose ML 2005. Anti-intercellular adhesion molecule-1 antibodies in sera of heart transplant recipients: A role in endothelial cell activation. Transplantation 80: 264–271 [DOI] [PubMed] [Google Scholar]
- Lazzeri E, Rotondi M, Mazzinghi B, Lasagni L, Buonamano A, Rosati A, Pradella F, Fossombroni V, La Villa G, Gacci M, et al. 2005. High CXCL10 expression in rejected kidneys and predictive role of pretransplant serum CXCL10 for acute rejection and chronic allograft nephropathy. Transplantation 79: 1215–1220 [DOI] [PubMed] [Google Scholar]
- Lee I, Wang L, Wells AD, Ye Q, Han R, Dorf ME, Kuziel WA, Rollins BJ, Chen L, Hancock WW 2003. Blocking the monocyte chemoattractant protein-1/CCR2 chemokine pathway induces permanent survival of islet allografts through a programmed death-1 ligand-1-dependent mechanism. J Immunol 171: 6929–6935 [DOI] [PubMed] [Google Scholar]
- Lee I, Wang L, Wells AD, Dorf ME, Ozkaynak E, Hancock W 2005. Recruitment of FoxP3+ T regulatory cells mediating allograft tolerance depends on the CCR4 chemokine receptor. J Exp Med 201: 1037–1044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lefaucheur C, Suberbielle-Boissel C, Hill GS, Nochy D, Andrade J, Antoine C, Gautreau C, Charron D, Glotz D 2008. Clinical relevance of preformed HLA donor-specific antibodies in kidney transplantation. Am J Transplant 8: 324–331 [DOI] [PubMed] [Google Scholar]
- Lefaucheur C, Nochy D, Andrade J, Verine J, Gautreau C, Charron D, Hill GS, Glotz D, Suberbielle-Boissel C 2009. Comparison of combination plasmapheresis/IVIg/anti-CD20 versus high-dose IVIg in the treatment of antibody-mediated rejection. Am J Transplant 9: 1099–1107 [DOI] [PubMed] [Google Scholar]
- Lefaucheur C, Loupy A, Hill GS, Andrade J, Nochy D, Antoine C, Gautreau C, Charron D, Glotz D, Suberbielle-Boissel C 2010. Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol 21: 1398–1406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Texier L, Thebault P, Lavault A, Usal C, Merieau E, Quillard T, Charreau B, Soulillou JP, Cuturi MC, Brouard S, et al. 2011. Long-term allograft tolerance is characterized by the accumulation of B cells exhibiting an inhibited profile. Am J Transplant 11: 429–438 [DOI] [PubMed] [Google Scholar]
- Li XC, Baldwin WM III 2012. NK cells: New partners in antibody-triggered chronic rejection. Am J Transplant 12: 275–276 [DOI] [PubMed] [Google Scholar]
- Li B, Hartono C, Ding R, Sharma VK, Ramaswamy R, Qian B, Serur D, Mouradian J, Schwartz JE, Suthanthiran M 2001. Noninvasive diagnosis of renal-allograft rejection by measurement of messenger RNA for perforin and granzyme B in urine. N Engl J Med 344: 947–954 [DOI] [PubMed] [Google Scholar]
- Li Y, Koshiba T, Yoshizawa A, Yonekawa Y, Masuda K, Ito A, Ueda M, Mori T, Kawamoto H, Tanaka Y, et al. 2004. Analyses of peripheral blood mononuclear cells in operational tolerance after pediatric living donor liver transplantation. Am J Transplant 4: 2118–2125 [DOI] [PubMed] [Google Scholar]
- Li J, Deane JA, Campanale NV, Bertram JF, Ricardo SD 2007. The contribution of bone marrow-derived cells to the development of renal interstitial fibrosis. Stem Cells 25: 697–706 [DOI] [PubMed] [Google Scholar]
- Li L, Chen A, Chaudhuri A, Kambham N, Sigdel T, Chen R, Sarwal MM 2010. Compartmental localization and clinical relevance of MICA antibodies after renal transplantation. Transplantation 89: 312–319 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Chen G, Ye P, Wang S, Zhang K, Chen W, Stepkowski SM, Li J, Zhong S, Xia J 2011. CCR5 blockade in combination with cyclosporine increased cardiac graft survival and generated alternatively activated macrophages in primates. J Immunol 186: 3753–3761 [DOI] [PubMed] [Google Scholar]
- Li H, Xie HY, Zhou L, Feng XW, Wang WL, Liang TB, Zhang M, Zheng SS 2012. Copy number variation in CCL3L1 gene is associated with susceptibility to acute rejection in patients after liver transplantation. Clin Transplant 26: 314–321 [DOI] [PubMed] [Google Scholar]
- Lin D, Lavender H, Soilleux EJ, O’Callaghan CA 2012. NF-κB regulates MICA gene transcription in endothelial cell through a genetically inhibitable control site. J Biol Chem 287: 4299–4310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindert S, Wickert L, Sawitza I, Wiercinska E, Gressner AM, Dooley S, Breitkopf K 2005. Transdifferentiation-dependent expression of α-SMA in hepatic stellate cells does not involve TGF-β pathways leading to coinduction of collagen type I and thrombospondin-2. Matrix Biol 24: 198–207 [DOI] [PubMed] [Google Scholar]
- Liu X, Mishra P, Yu S, Beckmann J, Wendland M, Kocks J, Seth S, Hoffmann K, Hoffmann M, Kremmer E, et al. 2011. Tolerance induction towards cardiac allografts under costimulation blockade is impaired in CCR7-deficient animals but can be restored by adoptive transfer of syngeneic plasmacytoid dendritic cells. Eur J Immunol 41: 611–623 [DOI] [PubMed] [Google Scholar]
- Lo DJ, Weaver TA, Kleiner DE, Mannon RB, Jacobson LM, Becker BN, Swanson SJ, Hale DA, Kirk AD 2011. Chemokines and their receptors in human renal allotransplantation. Transplantation 91: 70–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lombardi G, Sidhu S, Daly M, Batchelor JR, Makgoba W, Lechler RI 1990. Are primary alloresponses truly primary? Int Immunol 2: 9–13 [DOI] [PubMed] [Google Scholar]
- Lorenzen JM, Volkmann I, Fiedler J, Schmidt M, Scheffner I, Haller H, Gwinner W, Thum T 2011. Urinary miR-210 as a mediator of acute T-cell mediated rejection in renal allograft recipients. Am J Transplant 11: 2221–2227 [DOI] [PubMed] [Google Scholar]
- Lozano JJ, Pallier A, Martinez-Llordella M, Danger R, Lopez M, Giral M, Londono MC, Rimola A, Soulillou JP, Brouard S, et al. 2011. Comparison of transcriptional and blood cell-phenotypic markers between operationally tolerant liver and kidney recipients. Am J Transplant 11: 1916–1926 [DOI] [PubMed] [Google Scholar]
- Lu KC, Jaramillo A, Lecha RL, Schuessler RB, Aloush A, Trulock EP, Mendeloff EN, Huddleston CB, Alexander Patterson G, Mohanakumar T 2002. Interleukin-6 and interferon-γ gene polymorphisms in the development of bronchiolitis obliterans syndrome after lung transplantation. Transplantation 74: 1297–1302 [DOI] [PubMed] [Google Scholar]
- Lucas JG, Co JP, Nwaogwugwu UT, Dosani I, Sureshkumar KK 2011. Antibody-mediated rejection in kidney transplantation: An update. Expert Opin Pharmacother 12: 579–592 [DOI] [PubMed] [Google Scholar]
- Lumeng CN, Bodzin JL, Saltiel AR 2007. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175–184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacDermott RP 1996. Alterations of the mucosal immune system in inflammatory bowel disease. J Gastroenterol 31: 907–916 [DOI] [PubMed] [Google Scholar]
- Madsen CB, Norgaard A, Iversen M, Ryder LP 2010. Elevated mRNA levels of CTLA-4, FoxP3, and granzyme B in BAL, but not in blood, during acute rejection of lung allografts. Transpl Immunol 24: 26–32 [DOI] [PubMed] [Google Scholar]
- Magil AB 2005. Infiltrating cell types in transplant glomerulitis: Relationship to peritubular capillary C4d deposition. Am J Kidney Dis 45: 1084–1089 [DOI] [PubMed] [Google Scholar]
- Magil AB, Tinckam KJ 2006. Focal peritubular capillary C4d deposition in acute rejection. Nephrol Dial Transplant 21: 1382–1388 [DOI] [PubMed] [Google Scholar]
- Mahesh B, Leong HS, McCormack A, Sarathchandra P, Holder A, Rose ML 2007. Autoantibodies to vimentin cause accelerated rejection of cardiac allografts. Am J Pathol 170: 1415–1427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maier S, Tertilt C, Chambron N, Gerauer K, Huser N, Heidecke CD, Pfeffer K 2001. Inhibition of natural killer cells results in acceptance of cardiac allografts in CD28−/− mice. Nat Med 7: 557–562 [DOI] [PubMed] [Google Scholar]
- Manes TD, Pober JS 2008. Antigen presentation by human microvascular endothelial cells triggers ICAM-1-dependent transendothelial protrusion by, and fractalkine-dependent transendothelial migration of, effector memory CD4+ T cells. J Immunol 180: 8386–8392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manes TD, Shiao SL, Dengler TJ, Pober JS 2007. TCR signaling antagonizes rapid IP-10-mediated transendothelial migration of effector memory CD4+ T cells. J Immunol 178: 3237–3243 [DOI] [PubMed] [Google Scholar]
- Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M 2004. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25: 677–686 [DOI] [PubMed] [Google Scholar]
- Mao Q, Terasaki PI, Cai J, Briley K, Catrou P, Haisch C, Rebellato L 2007a. Extremely high association between appearance of HLA antibodies and failure of kidney grafts in a five-year longitudinal study. Am J Transplant 7: 864–871 [DOI] [PubMed] [Google Scholar]
- Mao Q, Terasaki PI, Cai J, El-Awar N, Rebellato L 2007b. Analysis of HLA class I specific antibodies in patients with failed allografts. Transplantation 83: 54–61 [DOI] [PubMed] [Google Scholar]
- Mao Y, Wang M, Zhou Q, Jin J, Wang Y, Peng W, Wu J, Shou Z, Chen J 2011. CXCL10 and CXCL13 Expression were highly up-regulated in peripheral blood mononuclear cells in acute rejection and poor response to anti-rejection therapy. J Clin Immunol 31: 414–418 [DOI] [PubMed] [Google Scholar]
- Martinez-Dolz L, Almenar L, Reganon E, Vila V, Sanchez-Soriano R, Martinez-Sales V, Moro J, Aguero J, Sanchez-Lazaro I, Salvador A 2009. What is the best biomarker for diagnosis of rejection in heart transplantation? Clin Transplant 23: 672–680 [DOI] [PubMed] [Google Scholar]
- Martinez-Llordella M, Puig-Pey I, Orlando G, Ramoni M, Tisone G, Rimola A, Lerut J, Latinne D, Margarit C, Bilbao I, et al. 2007. Multiparameter immune profiling of operational tolerance in liver transplantation. Am J Transplant 7: 309–319 [DOI] [PubMed] [Google Scholar]
- Martinez-Llordella M, Lozano JJ, Puig-Pey I, Orlando G, Tisone G, Lerut J, Benitez C, Pons JA, Parrilla P, Ramirez P, et al. 2008. Using transcriptional profiling to develop a diagnostic test of operational tolerance in liver transplant recipients. J Clin Invest 118: 2845–2857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matas AJ, Humar A, Gillingham KJ, Payne WD, Gruessner RW, Kandaswamy R, Dunn DL, Najarian JS, Sutherland DE 2002. Five preventable causes of kidney graft loss in the 1990s: A single-center analysis. Kidney Int 62: 704–714 [DOI] [PubMed] [Google Scholar]
- Matz M, Beyer J, Wunsch D, Mashreghi MF, Seiler M, Pratschke J, Babel N, Volk HD, Reinke P, Kotsch K 2006. Early post-transplant urinary IP-10 expression after kidney transplantation is predictive of short- and long-term graft function. Kidney Int 69: 1683–1690 [DOI] [PubMed] [Google Scholar]
- McDouall RM, Batten P, McCormack A, Yacoub MH, Rose ML 1997. MHC class II expression on human heart microvascular endothelial cells: Exquisite sensitivity to interferon-γ and natural killer cells. Transplantation 64: 1175–1180 [DOI] [PubMed] [Google Scholar]
- McNerney ME, Lee KM, Zhou P, Molinero L, Mashayekhi M, Guzior D, Sattar H, Kuppireddi S, Wang CR, Kumar V, Alegre ML 2006. Role of natural killer cell subsets in cardiac allograft rejection. Am J Transplant 6: 505–513 [DOI] [PubMed] [Google Scholar]
- Meier-Kriesche HU, Schold JD, Kaplan B 2004. Long-term renal allograft survival: Have we made significant progress or is it time to rethink our analytic and therapeutic strategies? Am J Transplant 4: 1289–1295 [DOI] [PubMed] [Google Scholar]
- Mengel M, Sis B, Haas M, Colvin RB, Halloran PF, Racusen LC, Solez K, Cendales L, Demetris AJ, Drachenberg CB, et al. 2012. Banff 2011 meeting report: New concepts in antibody-mediated rejection. Am J Transplant 12: 563–570 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer-ter-Vehn T, Sieprath S, Katzenberger B, Gebhardt S, Grehn F, Schlunck G 2006. Contractility as a prerequisite for TGF-β-induced myofibroblast transdifferentiation in human tenon fibroblasts. Invest Ophthalmol Vis Sci 47: 4895–4904 [DOI] [PubMed] [Google Scholar]
- Mezzano SA, Aros CA, Droguett A, Burgos ME, Ardiles LG, Flores CA, Carpio D, Vio CP, Ruiz-Ortega M, Egido J 2003. Renal angiotensin II up-regulation and myofibroblast activation in human membranous nephropathy. Kidney Int Suppl 86: S39–S45 [DOI] [PubMed] [Google Scholar]
- Miura M, El-Sawy T, Fairchild FL 2003. Neutrophils mediate parenchymal tissue necrosis and accelerate the rejection of complete major histocompatibility complex-disparate cardiac allografts in the absence of interferon-γ. Am J Pathol 162: 509–519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molesworth-Kenyon SJ, Oakes JE, Lausch RN 2005. A novel role for neutrophils as a source of T cell-recruiting chemokines IP-10 and Mig during the DTH response to HSV-1 antigen. J Leukoc Biol 77: 552–559 [DOI] [PubMed] [Google Scholar]
- Montgomery RA, Hardy MA, Jordan SC, Racusen LC, Ratner LE, Tyan DB, Zachary AA 2004. Consensus opinion from the antibody working group on the diagnosis, reporting, and risk assessment for antibody-mediated rejection and desensitization protocols. Transplantation 78: 181–185 [DOI] [PubMed] [Google Scholar]
- Montgomery RA, Lonze BE, King KE, Kraus ES, Kucirka LM, Locke JE, Warren DS, Simpkins CE, Dagher NN, Singer AL, et al. 2011. Desensitization in HLA-incompatible kidney recipients and survival. N Engl J Med 365: 318–326 [DOI] [PubMed] [Google Scholar]
- Montgomery JR, Berger JC, Warren DS, James NT, Montgomery RA, Segev DL 2012. Outcomes of ABO-incompatible kidney transplantation in the United States. Transplantation 93: 603–609 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mosser DM, Edwards JP 2008. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8: 958–969 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moxham VF, Karegli J, Phillips RE, Brown KL, Tapmeier TT, Hangartner R, Sacks SH, Wong W 2008. Homeostatic proliferation of lymphocytes results in augmented memory-like function and accelerated allograft rejection. J Immunol 180: 3910–3918 [DOI] [PubMed] [Google Scholar]
- Nadazdin O, Boskovic S, Murakami T, Tocco G, Smith RN, Colvin RB, Sachs DH, Allan J, Madsen JC, Kawai T, et al. 2011. Host alloreactive memory T cells influence tolerance to kidney allografts in nonhuman primates. Sci Transl Med 3: 86ra51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nafady-Hego H, Li Y, Ohe H, Zhao X, Satoda N, Sakaguchi S, Wood K, Uemoto S, Koshiba T 2010. The generation of donor-specific CD4+CD252+CD45RA+ naive regulatory T cells in operationally tolerant patients after pediatric living-donor liver transplantation. Transplantation 90: 1547–1555 [DOI] [PubMed] [Google Scholar]
- Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chapman JR 2003. The natural history of chronic allograft nephropathy. N Engl J Med 349: 2326–2333 [DOI] [PubMed] [Google Scholar]
- Nelsestuen GL, Martinez MB, Hertz MI, Savik K, Wendt CH 2005. Proteomic identification of human neutrophil α-defensins in chronic lung allograft rejection. Proteomics 5: 1705–1713 [DOI] [PubMed] [Google Scholar]
- Neujahr DC, Perez SD, Mohammed A, Ulukpo O, Lawrence EC, Fernandez F, Pickens A, Force SD, Song M, Larsen CP, et al. 2012. Cumulative exposure to γ interferon-dependent chemokines CXCL9 and CXCL10 correlates with worse outcome after lung transplant. Am J Transplant 12: 438–446 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, Burlingham WJ, Marks WH, Sanz I, Lechler RI, et al. 2010. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest 120: 1836–1847 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ng YH, Oberbarnscheidt MH, Chandramoorthy HC, Hoffman R, Chalasani G 2010. B cells help alloreactive T cells differentiate into memory T cells. Am J Transplant 10: 1970–1980 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nickeleit V, Zeiler M, Gudat F, Thiel G, Mihatsch MJ 2002. Detection of the complement degradation product C4d in renal allografts: Diagnostic and therapeutic implications. J Am Soc Nephrol 13: 242–251 [DOI] [PubMed] [Google Scholar]
- Niimi M, Hara M, Witzke O, Morris PJ, Wood KJ 1998. Donor resting B cells induce indefinite prolongation of fully allogeneic cardiac grafts when delivered with anti-immunoglobulin-D monoclonal antibody: Evidence for tolerogenicity of donor resting B cells in vivo. Transplantation 66: 1786–1792 [DOI] [PubMed] [Google Scholar]
- Noorchashm H, Reed AJ, Rostami SY, Mozaffari R, Zekavat G, Koeberlein B, Caton AJ, Naji A 2006. B cell-mediated antigen presentation is required for the pathogenesis of acute cardiac allograft rejection. J Immunol 177: 7715–7722 [DOI] [PubMed] [Google Scholar]
- Nozaki T, Amano H, Bickerstaff A, Orosz CG, Novick AC, Tanabe K, Fairchild RL 2007. Antibody-mediated rejection of cardiac allografts in CCR5-deficient recipients. J Immunol 179: 5238–5245 [DOI] [PubMed] [Google Scholar]
- Ochando JC, Krieger NR, Bromberg JS 2006. Direct versus indirect allorecognition: Visualization of dendritic cell distribution and interactions during rejection and tolerization. Am J Transplant 6: 2488–2496 [DOI] [PubMed] [Google Scholar]
- Ojo AO, Hanson JA, Wolfe RA, Leichtman AB, Agodoa LY, Port FK 2000. Long-term survival in renal transplant recipients with graft function. Kidney Int 57: 307–313 [DOI] [PubMed] [Google Scholar]
- Ozawa M, Terasaki PI, Lee JH, Castro R, Alberu J, Alonso C, Alvarez I, Toledo R, Alvez H, Monterio M, et al. 2007. 14th International HLA and Immunogenetics Workshop: Report on the prospective chronic rejection project. Tissue Antigens 69: 174–179 [DOI] [PubMed] [Google Scholar]
- Paantjens AW, van de Graaf EA, Kwakkel-van Erp JM, Hoefnagel T, van Kessel DA, van den Bosch JM, Otten HG 2011. Lung transplantation affects expression of the chemokine receptor type 4 on specific T cell subsets. Clin Exp Immunol 166: 103–109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pallier A, Hillion S, Danger R, Giral M, Racape M, Degauque N, Dugast E, Ashton-Chess J, Pettre S, Lozano JJ, et al. 2010. Patients with drug-free long-term graft function display increased numbers of peripheral B cells with a memory and inhibitory phenotype. Kidney Int 78: 503–513 [DOI] [PubMed] [Google Scholar]
- Panigrahi A, Siddiqui JA, Rai A, Margoob A, Khaira A, Bhowmik D, Tiwari SC, Guleria S, Mehra MK 2007. Allosensitization to HLA and MICA is an important measure of renal graft outcome. Clin Transpl 2007: 211–217 [PubMed] [Google Scholar]
- Pascual M, Theruvath T, Kawai T, Tolkoff-Rubin N, Cosimi AB 2002. Strategies to improve long-term outcomes after renal transplantation. N Engl J Med 346: 580–590 [DOI] [PubMed] [Google Scholar]
- Pearl JP, Parris J, Hale DA, Hoffmann SC, Bernstein WB, McCoy KL, Swanson SJ, Mannon RB, Roederer M, Kirk AD 2005. Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant 5: 465–474 [DOI] [PubMed] [Google Scholar]
- Pedagogos E, Hewitson TD, Walker RG, Nicholis KM, Becker GJ 1997. Myofibroblast involvement in chronic transplant rejection. Transplantation 64: 1192–1197 [DOI] [PubMed] [Google Scholar]
- Pei R, Lee JH, Shih NJ, Chen M, Terasaki PI 2003. Single human leukocyte antigen flow cytometry beads for accurate identification of human leukocyte antigen antibody specificities. Transplantation 75: 43–49 [DOI] [PubMed] [Google Scholar]
- Perrella O, Sbreglia C, Arenga G, Perrella A, Ferrara A, D’Antonio A, Di Costanzo G, Atripaldi L, Alone C, Sciano D, et al. 2006. Acute rejection after liver transplantation: Is there a specific immunological pattern? Transplant Proc 38: 3594–3596 [DOI] [PubMed] [Google Scholar]
- Pilmore HL, Painter DM, Bishop GA, McCaughan GW, Eris JM 2000. Early up-regulation of macrophages and myofibroblasts: A new marker for development of chronic renal allograft rejection. Transplantation 69: 2658–2662 [DOI] [PubMed] [Google Scholar]
- Pons JA, Revilla-Nuin B, Baroja-Mazo A, Ramirez P, Martinez-Alarcon L, Sanchez-Bueno F, Robles R, Rios A, Aparicio P, Parrilla P 2008. FoxP3 in peripheral blood is associated with operational tolerance in liver transplant patients during immunosuppression withdrawal. Transplantation 86: 1370–1378 [DOI] [PubMed] [Google Scholar]
- Port FK, Dykstra DM, Merion RM, Wolfe RA 2004. Organ donation and transplantation trends in the USA, 2003. Am J Transplant 4: 7–12 [DOI] [PubMed] [Google Scholar]
- Posselt AM, Vincenti F, Bedolli M, Lantz M, Roberts JP, Hirose R 2003. CD69 expression on peripheral CD8 T cells correlates with acute rejection in renal transplant recipients. Transplantation 76: 190–195 [DOI] [PubMed] [Google Scholar]
- Pratschke J, Stauch D, Kotsch K 2009. Role of NK and NKT cells in solid organ transplantation. Transpl Int 22: 859–868 [DOI] [PubMed] [Google Scholar]
- Qi F, Adair A, Ferenbach D, Vass DG, Mylonas KJ, Kipari T, Clay M, Kluth DC, Hughes J, Marson LP 2008. Depletion of cells of monocyte lineage prevents loss of renal microvasculature in murine kidney transplantation. Transplantation 86: 1267–1274 [DOI] [PubMed] [Google Scholar]
- Racusen LC, Solez K, Colvin RB, Bonsib SM, Castro MC, Cavallo T, Croker BP, Demetris AJ, Drachenberg CB, Fogo AB, et al. 1999. The Banff 97 working classification of renal allograft pathology. Kidney Int 55: 713–723 [DOI] [PubMed] [Google Scholar]
- Racusen LC, Colvin RB, Solez K, Mihatsch MJ, Halloran PF, Campbell PM, Cecka MJ, Cosyns JP, Demetris AJ, Fishbein MC, et al. 2003. Antibody-mediated rejection criteria—An addition to the Banff 97 classification of renal allograft rejection. Am J Transplant 3: 708–714 [DOI] [PubMed] [Google Scholar]
- Raedler H, Vieyra MB, Leisman S, Lakhani P, Kwan W, Yang M, Johnson K, Faas SJ, Tamburini P, Heeger PS 2011. Anti-complement component C5 mAb synergizes with CTLA4Ig to inhibit alloreactive T cells and prolong cardiac allograft survival in mice. Am J Transplant 11: 1397–1406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raffetseder U, Rauen T, Djudjaj S, Kretzler M, En-Nia A, Tacke F, Zimmermann HW, Nelson PJ, Frye BC, Floege J, et al. 2009. Differential regulation of chemokine CCL5 expression in monocytes/macrophages and renal cells by Y-box protein-1. Kidney Int 75: 185–196 [DOI] [PubMed] [Google Scholar]
- Ramirez AM, Shen Z, Ritzenthaler JD, Roman J 2006. Myofibroblast transdifferentiation in obliterative bronchiolitis: TGF-β signaling through smad3-dependent and independent pathways. Am J Transplant 6: 2080–2088 [DOI] [PubMed] [Google Scholar]
- Ramsperger-Gleixner M, Spriewald BM, Tandler R, Kondruweit M, Amann K, Weyand M, Ensminger SM 2011. Increased transcript levels of TNF-α, TGF-β, and granzyme B in endomyocardial biopsies correlate with allograft rejection. Exp Clin Transplant 9: 387–392 [PubMed] [Google Scholar]
- Rebellato LM, Ozawa M, Verbanac KM, Catrou P, Haisch CE, Terasaki PI 2006. Clinical and anti-HLA antibody profile of nine renal transplant recipients with failed grafts: Donor-specific and non-donor-specific antibody development. Clin Transpl 2006: 241–253 [PubMed] [Google Scholar]
- Regele H, Bohmig GA, Habicht A, Gollowitzer D, Schillinger M, Rockenschaub S, Watschinger B, Kerjaschki D, Exner M 2002. Capillary deposition of complement split product C4d in renal allografts is associated with basement membrane injury in peritubular and glomerular capillaries: A contribution of humoral immunity to chronic allograft rejection. J Am Soc Nephrol 13: 2371–2380 [DOI] [PubMed] [Google Scholar]
- Ridge JP, Di Rosa F, Matzinger P 1998. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature 393: 474–478 [DOI] [PubMed] [Google Scholar]
- Ro H, Hwang YH, Kim H, Jeong JC, Lee H, Doh YS, Park HC, Oh KH, Park MH, Ha J, et al. 2011. Association of polymorphisms of interleukin-8, CXCR1, CXCR2, and selectin with allograft outcomes in kidney transplantation. Transplantation 91: 57–64 [DOI] [PubMed] [Google Scholar]
- Rogers PR, Dubey C, Swain SL 2000. Qualitative changes accompany memory T cell generation: Faster, more effective responses at lower doses of antigen. J Immunol 164: 2338–2346 [DOI] [PubMed] [Google Scholar]
- Roos-van Groningen MC, Scholten EM, Lelieveld PM, Rowshani AT, Baelde HJ, Bajema IM, Florquin S, Bemelman FJ, de Heer E, de Fijter JW, et al. 2006. Molecular comparison of calcineurin inhibitor-induced fibrogenic responses in protocol renal transplant biopsies. J Am Soc Nephrol 17: 881–888 [DOI] [PubMed] [Google Scholar]
- Rosenblum JM, Shimoda N, Schenk AD, Zhang H, Kish DD, Keslar K, Farber JM, Fairchild RL 2010. CXC chemokine ligand CXCL9 and CXCL10 are antagonistic costimulation molecules during the priming of alloreactive T cell effectors. J Immunol 184: 3450–3460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossini M, Cheunsuchon B, Donnert E, Ma LJ, Thomas JW, Neilson EG, Fogo AB 2005. Immunolocalization of fibroblast growth factor-1 (FGF-1), its receptor (FGFR-1), and fibroblast-specific protein-1 (FSP-1) in inflammatory renal disease. Kidney Int 68: 2621–2628 [DOI] [PubMed] [Google Scholar]
- Rotondi M, Rosati A, Buonamano A, Lasagni L, Lazzeri E, Pradella F, Fossombroni V, Cirami C, Liotta F, La Villa G, et al. 2004. High pretransplant serum levels of CXCL10/IP-10 are related to increased risk of renal allograft failure. Am J Transplant 4: 1466–1474 [DOI] [PubMed] [Google Scholar]
- Rotondi M, Netti GS, Lazzeri E, Stallone G, Bertoni E, Chiovato L, Grandaliano G, Gesualdo L, Salvadori M, Schena FP, et al. 2010. High pretransplant serum levels of CXCL9 are associated with increased risk of acute rejection and graft failure in kidney graft recipients. Transpl Int 23: 465–475 [DOI] [PubMed] [Google Scholar]
- Sagoo P, Perucha E, Sawitzki B, Tomiuk S, Stephens DA, Miqueu P, Chapman S, Craciun L, Sergeant R, Brouard S, et al. 2010. Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest 120: 1848–1861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sagoo P, Ali N, Garg G, Nestle FO, Lechler RI, Lombardi G 2011. Human regulatory T cells with alloantigen specificity are more potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T cells. Sci Transl Med 3: 83ra42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salama AD, Womer KL, Sayegh MH 2007. Clinical transplantation tolerance: Many rivers to cross. J Immunol 178: 5419–5423 [DOI] [PubMed] [Google Scholar]
- Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A 1999. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401: 708–712 [DOI] [PubMed] [Google Scholar]
- Sarma NJ, Tiriveedhi V, Angaswamy N, Mohanakumar T 2012. Role of antibodies to self-antigens in chronic allograft rejection: Potential mechanism and therapeutic implications. Hum Immunol 10: 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaub S, Nickerson P, Rush D, Mayr M, Hess C, Golian M, Stefura W, Hayglass K 2009. Urinary CXCL9 and CXCL10 levels correlate with the extent of subclinical tubulitis. Am J Transplant 9: 1347–1353 [DOI] [PubMed] [Google Scholar]
- Schluns KS, Kieper WC, Jameson SC, Lefrancois L 2000. Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat Immunol 1: 426–432 [DOI] [PubMed] [Google Scholar]
- Schowengerdt KO, Fricker FJ, Bahjat KS, Kuntz ST 2000. Increased expression of the lymphocyte early activation marker CD69 in peripheral blood correlates with histologic evidence of cardiac allograft rejection. Transplantation 69: 2102–2107 [DOI] [PubMed] [Google Scholar]
- Seiler M, Brabcova I, Viklicky O, Hribova P, Rosenberger C, Pratschke J, Lodererova A, Matz M, Schonemann C, Reinke P, et al. 2007. Heightened expression of the cytotoxicity receptor NKG2D correlates with acute and chronic nephropathy after kidney transplantation. Am J Transplant 7: 423–433 [DOI] [PubMed] [Google Scholar]
- Shenoy KV, Solomides C, Cordova F, Rogers TJ, Ciccolella D, Criner GJ 2012. Low CD4/CD8 ratio in bronchus-associated lymphoid tissue is associated with lung allograft rejection. J Transplant 2012: 928081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shevach EM 2009. Mechanisms of FoxP3+ T regulatory cell-mediated suppression. Immunity 30: 636–645 [DOI] [PubMed] [Google Scholar]
- Shin GT, Kim SJ, Lee TS, Oh CK, Kim H 2005. Gene expression of perforin by peripheral blood lymphocytes as a marker of acute rejection. Nephron Clin Pract 100: c63–c70 [DOI] [PubMed] [Google Scholar]
- Silva HM, Takenaka MC, Moraes-Vieira PM, Monteiro SM, Hernandez MO, Chaara W, Six A, Agena F, Sesterheim P, Barbe-Tuana FM, et al. 2012. Preserving the B-cell compartment favors operational tolerance in human renal transplantation. Mol Med 18: 733–743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sis B, Campbell PM, Mueller T, Hunter C, Cockfield SM, Cruz J, Meng C, Wishart D, Solez K, Halloran PF 2007. Transplant glomerulopathy, late antibody-mediated rejection and the ABCD tetrad in kidney allograft biopsies for cause. Am J Transplant 7: 1743–1752 [DOI] [PubMed] [Google Scholar]
- Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF 2009. Endothelial gene expression in kidney transplants with alloantibody indicates antibody-mediated damage despite lack of C4d staining. Am J Transplant 9: 2312–2323 [DOI] [PubMed] [Google Scholar]
- Smith C, Miles JJ, Khanna R 2012. Advances in direct T-cell alloreactivity: Function, avidity, biophysics and structure. Am J Transplant 12: 15–26 [DOI] [PubMed] [Google Scholar]
- Smyth LA, Herrera OB, Golshayan D, Lombardi G, Lechler RI 2006. A novel pathway of antigen presentation by dendritic and endothelial cells: Implications for allorecognition and infectious diseases. Transplantation 82: S15–S18 [DOI] [PubMed] [Google Scholar]
- Solez K, Vincenti F, Filo RS 1998. Histopathologic findings from 2-year protocol biopsies from a U.S. multicenter kidney transplant trial comparing tarolimus versus cyclosporine: A report of the FK506 Kidney Transplant Study Group. Transplantation 66: 1736–1740 [DOI] [PubMed] [Google Scholar]
- Solez K, Colvin RB, Racusen LC, Sis B, Halloran PF, Birk PE, Campbell PM, Cascalho M, Collins AB, Demetris AJ, et al. 2007. Banff ’05 meeting report: Differential diagnosis of chronic allograft injury and elimination of chronic allograft nephropathy (“CAN”). Am J Transplant 7: 518–526 [DOI] [PubMed] [Google Scholar]
- Solez K, Colvin RB, Racusen LC, Haas M, Sis B, Mengel M, Halloran PF, Baldwin W, Banfi G, Collins AB, et al. 2008. Banff 07 classification of renal allograft pathology: Updates and future directions. Am J Transplant 8: 753–760 [DOI] [PubMed] [Google Scholar]
- Sommer M, Gerth J, Stein G, Wolf G 2005. Transdifferentiation of endothelial and renal tubular epithelial cells into myofibroblast-like cells under in vitro conditions: A morphological analysis. Cells Tissues Organs 180: 204–214 [DOI] [PubMed] [Google Scholar]
- Sorrentino C, Scarinci A, D’Antuono T, Piccirilli M, Di Nicola M, Pasquale M, Di Iorio C, Di Carlo E 2006. Endomyocardial infiltration by B and NK cells foreshadows the recurrence of cardiac allograft rejection. J Pathol 209: 400–410 [DOI] [PubMed] [Google Scholar]
- Sumitran-Holgersson S 2008. Relevance of MICA and other non-HLA antibodies in clinical transplantation. Curr Opin Immunol 20: 607–613 [DOI] [PubMed] [Google Scholar]
- Sumitran-Holgersson S, Wilczek HE, Holgersson J, Soderstrom K 2002. Identification of the nonclassical HLA molecules, mica, as targets for humoral immunity associated with irreversible rejection of kidney allografts. Transplantation 74: 268–277 [DOI] [PubMed] [Google Scholar]
- Summers AM, Coupes BM, Brennan MF, Ralph SA, Short CD, Brenchley PE 2005. VEGF-460 genotype plays an important role in progression to chronic kidney disease stage 5. Nephrol Dial Transplant 20: 2427–2432 [DOI] [PubMed] [Google Scholar]
- Sun HJ, Zhou T, Wang Y, Fu YW, Jiang YP, Zhang LH, Zhang CB, Zhou HL, Gao BS, Shi YA, et al. 2011. Macrophages and T lymphocytes are the predominant cells in intimal arteritis of resected renal allografts undergoing acute rejection. Transpl Immunol 25: 42–48 [DOI] [PubMed] [Google Scholar]
- Surquin M, Le Moine A, Flamand V, Rombaut K, Demoor FX, Salmon I, Goldman M, Abramowicz D 2005. IL-4 deficiency prevents eosinophilic rejection and uncovers a role for neutrophils in the rejection of MHC class II disparate skin grafts. Transplantation 80: 1485–1492 [DOI] [PubMed] [Google Scholar]
- Taflin C, Favier B, Baudhuin J, Savenay A, Hemon P, Bensussan A, Charron D, Glotz D, Mooney N 2011. Human endothelial cells generate Th17 and regulatory T cells under inflammatory conditions. Proc Natl Acad Sci 108: 2891–2896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taflin C, Charron D, Glotz D, Mooney N 2012. Regulation of the CD4+ T cell allo-immune response by endothelial cells. Hum Immunol 16: 16. [DOI] [PubMed] [Google Scholar]
- Takeiri M, Tachibana M, Kaneda A, Ito A, Ishikawa Y, Nishiyama S, Goto R, Yamashita K, Shibasaki S, Hirokata G, et al. 2011. Inhibition of macrophage activation and suppression of graft rejection by DTCM-glutarimide, a novel piperidine derived from the antibiotic 9-methylstreptimidone. Inflamm Res 60: 879–888 [DOI] [PubMed] [Google Scholar]
- Tanaka K, Ozawa K, Teramukai S, Takada Y, Egawa H, Kaihara S, Fujimoto Y, Ogura Y, Kasahara M, Ono M, et al. 2006. Classification of human liver transplant recipients by their preoperative CD8+ T cell subpopulation and its relation to outcome. Liver Transpl 12: 792–800 [DOI] [PubMed] [Google Scholar]
- Tatapudi RR, Muthukumar T, Dadhania D, Ding R, Li B, Sharma VK, Lozada-Pastorio E, Seetharamu N, Hartono C, Serur D, et al. 2004. Noninvasive detection of renal allograft inflammation by measurements of mRNA for IP-10 and CXCR3 in urine. Kidney Int 65: 2390–2397 [DOI] [PubMed] [Google Scholar]
- Terasaki PI 2003. Humoral theory of transplantation. Am J Transplant 3: 665–673 [DOI] [PubMed] [Google Scholar]
- Terasaki PI, Cai J 2005. Humoral theory of transplantation: Further evidence. Curr Opin Immunol 17: 541–545 [DOI] [PubMed] [Google Scholar]
- Terasaki PI, Ozawa M 2004. Predicting kidney graft failure by HLA antibodies: A prospective trial. Am J Transplant 4: 438–443 [DOI] [PubMed] [Google Scholar]
- Thaunat O, Nicoletti A 2008. Lymphoid neogenesis in chronic rejection. Curr Opin Organ Transplant 13: 16–19 [DOI] [PubMed] [Google Scholar]
- Thaunat O, Field AC, Dai J, Louedec L, Patey N, Bloch MF, Mandet C, Belair MF, Bruneval P, Meilhac O, et al. 2005. Lymphoid neogenesis in chronic rejection: Evidence for a local humoral alloimmune response. Proc Natl Acad Sci 102: 14723–14728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thaunat O, Patey N, Morelon E, Michel JB, Nicoletti A 2006. Lymphoid neogenesis in chronic rejection: The murderer is in the house. Curr Opin Immunol 18: 576–579 [DOI] [PubMed] [Google Scholar]
- Thaunat O, Patey N, Gautreau C, Lechaton S, Fremeaux-Bacchi V, Dieu-Nosjean MC, Cassuto-Viguier E, Legendre C, Delahousse M, Lang P, et al. 2008. B cell survival in intragraft tertiary lymphoid organs after rituximab therapy. Transplantation 85: 1648–1653 [DOI] [PubMed] [Google Scholar]
- Thaunat O, Graff-Dubois S, Brouard S, Gautreau C, Varthaman A, Fabien N, Field AC, Louedec L, Dai J, Joly E, et al. 2010. Immune responses elicited in tertiary lymphoid tissues display distinctive features. PLoS ONE 5: e11398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thaunat O, Graff-Dubois S, Fabien N, Duthey A, Attuil-Audenis V, Nicoletti A, Patey N, Morelon E 2012. A stepwise breakdown of B-cell tolerance occurs within renal allografts during chronic rejection. Kidney Int 81: 207–219 [DOI] [PubMed] [Google Scholar]
- Tyden G, Genberg H, Tollemar J, Ekberg H, Persson NH, Tufveson G, Wadstrom J, Gabel M, Mjornstedt L 2009. A randomized, doubleblind, placebo-controlled, study of single-dose rituximab as induction in renal transplantation. Transplantation 87: 1325–1329 [DOI] [PubMed] [Google Scholar]
- Uppaluri R, Sheehan KC, Wang L, Bui JD, Brotman JJ, Lu B, Gerard C, Hancock WW, Schreiber RD 2008. Prolongation of cardiac and islet allograft survival by a blocking hamster anti-mouse CXCR3 monoclonal antibody. Transplantation 86: 137–147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valujskikh A, Pantenburg B, Heeger PS 2002. Primed allospecific T cells prevent the effects of costimulatory blockade on prolonged cardiac allograft survival in mice. Am J Transplant 2: 501–509 [DOI] [PubMed] [Google Scholar]
- Van der Touw W, Burrell B, Lal G, Bromberg JS 2012. NK cells are required for costimulatory blockade induced tolerance to vascularized allografts. Transplantation 20: 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veiga-Fernandes H, Walter U, Bourgeois C, McLean A, Rocha B 2000. Response of naive and memory CD8+ T cells to antigen stimulation in vivo. Nat Immunol 1: 47–53 [DOI] [PubMed] [Google Scholar]
- Wang Y, Dai H, Liu Z, Cheng X, Tellides G, Dai Z 2006. Neutralizing IL-7 promotes long-term allograft survival induced by CD40/CD40L costimulatory blockade. Am J Transplant 6: 2851–2860 [DOI] [PubMed] [Google Scholar]
- Wang S, Li J, Xie A, Wang G, Xia N, Ye P, Rui L, Xia J 2011. Dynamic changes in Th1, Th17, and FoxP3+ T cells in patients with acute cellular rejection after cardiac transplantation. Clin Transplant 25: E177–E186 [DOI] [PubMed] [Google Scholar]
- Wasowska BA, Qian Z, Cangello DL, Behrens E, Van Tran K, Layton J, Sanfilippo F, Baldwin MW III 2001. Passive transfer of alloantibodies restores acute cardiac rejection in IgKO mice. Transplantation 71: 727–736 [DOI] [PubMed] [Google Scholar]
- Won DI, Jeong HD, Kim YL, Suh JS 2006. Simultaneous detection of antibody binding and cytotoxicity in flow cytometry crossmatch for renal transplantation. Cytometry B Clin Cytom 70: 82–90 [DOI] [PubMed] [Google Scholar]
- Wu Z, Bensinger SJ, Zhang J, Chen C, Yuan X, Huang X, Markmann JF, Kassaee A, Rosengard BR, Hancock WW, et al. 2004. Homeostatic proliferation is a barrier to transplantation tolerance. Nat Med 10: 87–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu T, Bond G, Martin D, Nalesnik MA, Demetris AJ, Abu-Elmagd K 2006. Histopathologic characteristics of human intestine allograft acute rejection in patients pretreated with thymoglobulin or alemtuzumab. Am J Gastroenterol 101: 1617–1624 [DOI] [PubMed] [Google Scholar]
- Wynn TA 2008. Cellular and molecular mechanisms of fibrosis. J Pathol 214: 199–210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, Zhang X, Mannon RB, Kirk AD 2006. Platelet-derived or soluble CD154 induces vascularized allograft rejection independent of cell-bound CD154. J Clin Invest 116: 769–774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yabu JM, Higgins JP, Chen G, Sequeira F, Busque S, Tyan DB 2011. C1q-fixing human leukocyte antigen antibodies are specific for predicting transplant glomerulopathy and late graft failure after kidney transplantation. Transplantation 91: 342–347 [DOI] [PubMed] [Google Scholar]
- Yamada A, Salama AD, Sho M, Najafian N, Ito T, Forman JP, Kewalramani R, Sandner S, Harada H, Clarkson MR, et al. 2005. CD70 signaling is critical for CD28-independent CD8+ T cell-mediated alloimmune responses in vivo. J Immunol 174: 1357–1364 [DOI] [PubMed] [Google Scholar]
- Yang WS, Han NJ, Kim CS, Ahn H, Lee SK, Lee KU, Park SK 2005. STAT1-independent down-regulation of interferon-γ-induced class II transactivator and HLA-DR expression by transforming growth factor β-1 in human glomerular endothelial cells. Nephron Exp Nephrol 100: e124–e131 [DOI] [PubMed] [Google Scholar]
- Yoo S, Lee EY, Huh KH, Kim MS, Kim YS, Kim HO 2012. Role of plasma exchange in ABO-incompatible kidney transplantation. Ann Lab Med 32: 283–288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan X, Dong VM, Coito AJ, Waaga AM, Salama AD, Benjamin CD, Sayegh MH, Chandraker A 2002. A novel CD154 monoclonal antibody in acute and chronic rat vascularized cardiac allograft rejection. Transplantation 73: 1736–1742 [DOI] [PubMed] [Google Scholar]
- Yuan X, Paez-Cortez J, Schmitt-Knosalla I, D’Addio F, Mfarrej B, Donnarumma M, Habicht A, Clarkson MR, Iacomini J, Glimcher LH, et al. 2008. A novel role of CD4 Th17 cells in mediating cardiac allograft rejection and vasculopathy. J Exp Med 205: 3133–3144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan X, Ansari MJ, D’Addio F, Paez-Cortez J, Schmitt I, Donnarumma M, Boenisch O, Zhao X, Popoola J, Clarkson MR, et al. 2009. Targeting Tim-1 to overcome resistance to transplantation tolerance mediated by CD8 T17 cells. Proc Natl Acad Sci 106: 10734–10739 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zafar MN, Terasaki PI, Naqvi SA, Rizvi SA 2006. Non-HLA antibodies after rejection of HLA identical kidney transplants. Clin Transpl 2006: 421–426 [PubMed] [Google Scholar]
- Zarkhin V, Chalasani G, Sarwal MM 2010. The yin and yang of B cells in graft rejection and tolerance. Transplant Rev (Orlando) 24: 67–78 [DOI] [PubMed] [Google Scholar]
- Zhai Y, Meng L, Gao F, Busuttil RW, Kupiec-Weglinski JW 2002. Allograft rejection by primed/memory CD8+ T cells is CD154 blockade resistant: Therapeutic implications for sensitized transplant recipients. J Immunol 169: 4667–4673 [DOI] [PubMed] [Google Scholar]
- Zhai Y, Shen XD, Hancock WW, Gao F, Qiao B, Lassman C, Belperio JA, Strieter RM, Busuttil RW, Kupiec-Weglinski JW 2006. CXCR3+CD4+ T cells mediate innate immune function in the pathophysiology of liver ischemia/reperfusion injury. J Immunol 176: 6313–6322 [DOI] [PubMed] [Google Scholar]
- Zhang ZX, Wang S, Huang X, Min WP, Sun H, Liu W, Garcia B, Jevnikar AM 2008. NK cells induce apoptosis in tubular epithelial cells and contribute to renal ischemia-reperfusion injury. J Immunol 181: 7489–7498 [DOI] [PubMed] [Google Scholar]
- Zhou W, Farrar CA, Abe K, Pratt JR, Marsh JE, Wang Y, Stahl GL, Sacks SH 2000. Predominant role for C5b-9 in renal ischemia/reperfusion injury. J Clin Invest 105: 1363–1371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhuo M, Fujiki M, Wang M, Piard-Ruster K, Wai LE, Wei L, Martinez OM, Krams SM 2010. Identification of the rat NKG2D ligands, RAE1L and RRLT, and their role in allograft rejection. Eur J Immunol 40: 1748–1757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou Y, Stastny P, Susal C, Dohler B, Opelz G 2007. Antibodies against MICA antigens and kidney-transplant rejection. N Engl J Med 357: 1293–1300 [DOI] [PubMed] [Google Scholar]