Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 18(2): 289-292, 2018 02.
Article in English | MEDLINE | ID: mdl-28722285

ABSTRACT

Innate recognition of microbial products and danger molecules by monocytes and macrophages has been well established; this is mediated primarily by pattern-recognition receptors and is central to the activation of innate and adaptive immune cells required for productive immunity. Whether monocytes and macrophages are equipped with an allorecognition system that allows them to respond directly to allogeneic grafts is a topic of much debate. Recent studies provide compelling evidence that these cells can recognize allogeneic entities and that they mediate graft rejection via direct cytotoxicity and priming of alloreactive T cells. In addition, these studies have uncovered a mechanism of innate allorecognition based on detection of the polymorphic molecule signal regulatory protein α (SIRPα) on donor cells. Further understanding of innate allorecognition and its consequences would provide essential insight into allograft rejection and lead to better therapies for transplant patients.


Subject(s)
Graft Rejection/immunology , Immunity, Innate/immunology , Isoantigens/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Humans , Organ Transplantation , Transplantation Immunology
2.
Transpl Immunol ; 29(1-4): 88-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24120957

ABSTRACT

Repopulation of memory T cells (Tmem) in allograft recipients after lymphodepletion is a major barrier to transplant tolerance induction. Ineffective depletion of naïve T cells (Tn) and Tmem may predispose to repopulation of Tmem after transplantation. Cynomolgus macaque monkeys given heart allografts were lymphodepleted using Alemtuzumab (Campath-1H; anti-CD52). Peripheral blood (PB) and lymph nodes (LN) were analyzed for CD95(-) (Tn) and CD95(+) cells (Tmem), one day, one month and up to three months after Alemtuzumab infusion. CD52 expression, susceptibility to Alemtuzumab cytotoxicity and pro-apoptotic caspase-3 were evaluated in Tn and Tmem. In vivo, Alemtuzumab induction profoundly depleted lymphocytes in PB (99% reduction) but exerted a lesser effect in LN (70% reduction), with similar depletion of Tn and Tmem subsets. After transplantation, Tmem comprised the majority of lymphocytes in PB and LN. In vitro, LN T cells were more resistant to Alemtuzumab-mediated cytotoxicity than PB lymphocytes. CD4(+) Tn and Tmem were equally susceptible to Alemtuzumab-mediated cytotoxicity, whereas CD8(+) Tn were more resistant than CD8(+) Tmem. However, no significant differences in CD52 expression between lymphocyte subsets in PB and LN were observed. Caspase-3 expression was higher in PB than LN T cells. CD4(+) and CD8(+) Tn expressed lower levels of Caspase-3 than Tmem, in both PB and LN. Thus, after Alemtuzumab infusion, residual Tn in secondary lymphoid tissue may predispose to rapid recovery of Tmem in allograft recipients.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Heart Transplantation , Lymphocyte Depletion , Lymphoid Tissue/immunology , Memory, Short-Term/drug effects , Alemtuzumab , Allografts , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Caspase 3/immunology , Lymphoid Tissue/pathology , Macaca fascicularis , fas Receptor/immunology
3.
Am J Transplant ; 13(8): 1989-2005, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23758811

ABSTRACT

We examined the influence of regulatory dendritic cells (DCreg), generated from cytokine-mobilized donor blood monocytes in vitamin D3 and IL-10, on renal allograft survival in a clinically relevant rhesus macaque model. DCreg expressed low MHC class II and costimulatory molecules, but comparatively high levels of programmed death ligand-1 (B7-H1), and were resistant to pro-inflammatory cytokine-induced maturation. They were infused intravenously (3.5-10 × 10(6) /kg), together with the B7-CD28 costimulation blocking agent CTLA4Ig, 7 days before renal transplantation. CTLA4Ig was given for up to 8 weeks and rapamycin, started on Day -2, was maintained with tapering of blood levels until full withdrawal at 6 months. Median graft survival time was 39.5 days in control monkeys (no DC infusion; n = 6) and 113.5 days (p < 0.05) in DCreg-treated animals (n = 6). No adverse events were associated with DCreg infusion, and there was no evidence of induction of host sensitization based on circulating donor-specific alloantibody levels. Immunologic monitoring also revealed regulation of donor-reactive memory CD95(+) T cells and reduced memory/regulatory T cell ratios in DCreg-treated monkeys compared with controls. Termination allograft histology showed moderate combined T cell- and Ab-mediated rejection in both groups. These findings justify further preclinical evaluation of DCreg therapy and their therapeutic potential in organ transplantation.


Subject(s)
Dendritic Cells/transplantation , Graft Survival/immunology , Immune Tolerance/immunology , Immunologic Memory/immunology , Kidney Diseases/prevention & control , Kidney Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Abatacept , Animals , Combined Modality Therapy , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunoconjugates/immunology , Immunosuppressive Agents/therapeutic use , Kidney Diseases/immunology , Macaca mulatta , Male , Sirolimus/therapeutic use , Transplantation, Homologous
4.
Am J Transplant ; 13(3): 580-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23311531

ABSTRACT

Inbreeding depression and lack of genetic diversity in inbred mice could mask unappreciated causes of graft failure or remove barriers to tolerance induction. To test these possibilities, we performed heart transplantation between outbred or inbred mice. Unlike untreated inbred mice in which all allografts were rejected acutely (6-16 days posttransplantation), untreated outbred mice had heterogeneous outcomes, with grafts failing early (<4 days posttransplantation), acutely (6-24 days) or undergoing chronic rejection (>75 days). Blocking T cell costimulation induced long-term graft acceptance in both inbred and outbred mice, but did not prevent the early graft failure observed in the latter. Further investigation of this early phenotype established that it is dependent on the donor, and not the recipient, being outbred and that it is characterized by hemorrhagic necrosis and neutrophilic vasculitis in the graft without preformed, high titer antidonor antibodies in the recipient. Complement or neutrophil depletion prevented early failure of outbred grafts, whereas transplanting CD73-deficient inbred hearts, which are highly susceptible to ischemia-reperfusion injury, recapitulated the early phenotype. Therefore, outbred mice could provide broader insight into donor and recipient determinants of allograft outcomes but their hybrid vigor and genetic diversity do not constitute a uniform barrier to tolerance induction.


Subject(s)
Breeding , Graft Rejection/immunology , Graft Survival/physiology , Heart Transplantation , Immune Tolerance/immunology , Reperfusion Injury/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Genetic Fitness/physiology , Graft Rejection/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reperfusion Injury/mortality , Survival Rate , Transplantation, Homologous
5.
Am J Transplant ; 11(10): 2067-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21834913

ABSTRACT

Graft outcomes after kidney transplantation continue to be adversely affected by ischemia-reperfusion injury and rejection. High-resolution, real-time imaging of the transplanted kidney could shed valuable insights into these dynamic processes, but such methodology has not been established. Here we describe a technique for intravital imaging of the transplanted mouse kidney using multiphoton fluorescence microscopy. The technique enabled real-time, high-resolution imaging and quantitation of renal filtration, cell death, leukocyte adhesion and capillary blood flow after transplantation. Using this technique, we found that brief graft ischemia associated with the transplantation procedure led to a rapid decline in renal filtration accompanied by a significant increase in microvascular leakage and renal tubular epithelial cell death within the first 3 h after transplantation. No significant changes in leukocyte adhesion or capillary blood flow were observed during the same time period. This report establishes multiphoton fluorescence microscopy as a sensitive tool for simultaneously studying functional and structural perturbations that occur in the mouse kidney after transplantation and for investigating the migration of leukocytes to the graft.


Subject(s)
Kidney Transplantation , Kidney/pathology , Vitreous Body , Animals , Kidney/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
6.
Am J Transplant ; 10(4): 773-783, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20420638

ABSTRACT

As the target CD52 molecule is expressed on erythrocytes of most nonhuman primate strains, using alemtuzumab in these species would cause massive hemolysis. Six cynomolgus monkeys of Indonesian origin, screened by agglutination assay for absence of CD52 on erythrocytes, were administered alemtuzumab in a cumulative dose to a maximum of 60 mg/kg. In two monkeys, mycophenolate mofetil (MMF) was added as maintenance therapy. Complete depletion of T and B lymphocytes (>99.5%) was achieved with 20 mg/kg alemtuzumab and was more profound than in monkeys treated with antithymocyte globulin (n = 5), as quantified by flow cytometry. Repopulation was suppressed by weekly injections of 10 mg/kg. Without MMF, repopulation of CD20(+)B cells and CD8(+)T cells was complete within 2 and 3 months, respectively, and repopulation of CD4(+)T cells was 67% after 1 year. MMF significantly delayed CD4(+)T-cell repopulation. Among repopulating CD4(+) and CD8(+) T cells, a phenotypic shift was observed from CD45RA(hi)CD62L(hi) naïve cells toward CD45RA(lo)CD62L(lo) effector memory cells. In lymph nodes, the depletion of naïve cells was more profound than of memory cells, which may have initiated a proliferation of memory cells. This model offers opportunities to investigate lymphocyte depletion/repopulation phenomena, as well as the efficacy of alemtuzumab in preclinical transplantation models.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Cell Division/drug effects , Lymphocyte Depletion , Lymphocytes/cytology , Alemtuzumab , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/pharmacology , Antigens, CD/immunology , Flow Cytometry , Immunophenotyping , Lymphocytes/immunology , Macaca fascicularis
7.
Am J Transplant ; 10(1): 162-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19951284

ABSTRACT

Type I interferons (IFN-I) link innate to adaptive immunity in microbial infection, autoimmune disease and tumor immunity. It is not known whether IFN-I have an equally central role in alloimmunity. Here we tested this possibility by studying skin allograft survival and donor-specific CD8+ T-cell responses in mice that lack the IFN-I receptor (IFN-IR-/-). We found that IFN-IR-/- mice reject fully allogeneic wild-type skin grafts at the same rate as wild-type recipients. Similarly, allograft rejection was not delayed if IFN-IR-/- male skin was transplanted to syngeneic IFN-IR-/- female mice. Quantitation of the male (H-Y)-specific CD8+ T-cell response in these mice revealed normal generation of donor-specific CD8+ effector T cells but fourfold reduction in CD8+ memory T cells. Memory CD8+ T cells generated in the absence of IFN-IR had normal phenotype and recall function, assessed by ex vivo cytokine production and the ability of IFN-IR-/- mice to mount second set rejection. Finally, these memory T cells were maintained at a constant number despite their inability to respond to IFN-1. Our findings indicate that IFN-I cytokines are not critical for acute allograft rejection or for the expansion and differentiation of donor-specific CD8+ T cells into long-lived, functional memory T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Interferon Type I/immunology , Skin Transplantation/immunology , Animals , Female , Graft Rejection/etiology , Immunologic Memory , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Transplantation, Homologous
8.
Am J Transplant ; 9(12): 2716-26, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19845582

ABSTRACT

Xenotransplantation of porcine islets into diabetic non-human primates is characterized by (i) an initial massive graft loss possibly due to the instant blood-mediated inflammatory reaction and (ii) the requirement of intensive, clinically unfriendly immunosuppressive therapy. We investigated whether the transgenic expression of a human complement-regulatory protein (hCD46) on porcine islets would improve the outcome of islet xenotransplantation in streptozotocin-induced diabetic Cynomolgus monkeys. Immunosuppression consisted of thymoglobulin, anti-CD154 mAb for costimulation blockade, and mycophenolate mofetil. Following the transplantation of islets from wild-type pigs (n = 2) or from 1,3-galactosyltransferase gene-knockout pigs (n = 2), islets survived for a maximum of only 46 days, as evidenced by return to hyperglycemia and the need for exogenous insulin therapy. The transplantation of islets from hCD46 pigs resulted in graft survival and insulin-independent normoglycemia in four of five monkeys for the 3 months follow-up of the experiment. One normalized recipient, selected at random, was followed for >12 months. Inhibition of complement activation by the expression of hCD46 on the pig islets did not substantially reduce the initial loss of islet mass, rather was effective in limiting antibody-mediated rejection. This resulted in a reduced need for immunosuppression to preserve a sufficient islet mass to maintain normoglycemia long-term.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/physiopathology , Islets of Langerhans Transplantation/methods , Membrane Cofactor Protein/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Diabetes Mellitus, Experimental/surgery , Female , Macaca fascicularis , Male , Swine
9.
Am J Transplant ; 9(11): 2485-96, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19775318

ABSTRACT

The results of transplantation of human donor islets into the portal vein (PV) in patients with diabetes are encouraging. However, there are complications, for example, hemorrhage, thrombosis and an immediate loss of islets through the 'instant blood-mediated inflammatory reaction' (IBMIR). The gastric submucosal space (GSMS) offers potential advantages. Islets were isolated from adult pigs. Recipient pigs were made diabetic by streptozotocin. Donor islets were injected into the GSMS through a laparotomy (Group 1A, n = 4) or endoscopically (Group 1B, n = 8) or into the PV through a laparotomy (Group 2, n = 3). The pigs were followed for a maximum of 28 days. Monitoring of C-peptide in Group 1 indicated that there was minimal immediate loss of islets whereas in Group 2 there was considerable loss from IBMIR. In Group 1, there were significant reductions in mean blood glucose and mean exogenous insulin requirement between pretransplantation and 20 days posttransplantation. In Group 2, there was no significant reduction in either parameter. Insulin-positive cells were seen in the GSMS in Group 1, but not in the liver in Group 2. Endoscopic gastric submucosal transplantation of islets (ENDO-STI) offers a minimally invasive and quick approach to islet transplantation, avoids IBMIR and warrants further exploration.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Endoscopy/methods , Gastric Mucosa/surgery , Islets of Langerhans Transplantation/methods , Animals , Blood Glucose/metabolism , C-Peptide/blood , Combined Modality Therapy , Diabetes Mellitus, Experimental/drug therapy , Female , Graft Rejection/drug therapy , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival , Hypoglycemic Agents/pharmacology , Immunosuppressive Agents/pharmacology , Insulin/pharmacology , Islets of Langerhans Transplantation/immunology , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Pancreatectomy , Sus scrofa , Tacrolimus/pharmacology , Transplantation, Homologous
10.
Am J Transplant ; 9(9): 2057-66, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19624567

ABSTRACT

T-cell alloimmunity plays a dominant role in allograft rejection. The precise contribution of naïve and memory T cells to this response however remains unclear. To address this question, we established an ex vivo flow-cytometric assay that simultaneously measures proliferation, precursor frequency and effector molecule (IFNgamma, granzyme B/perforin) production of alloreactive T cells. By applying this assay to peripheral blood mononuclear cells from healthy volunteers, we demonstrate that the CD4+ and CD8+ populations mount similar proliferative responses and contain comparable frequencies of alloreactive precursors. Effector molecule expression, however, was significantly higher among CD8+ T cells. Analysis of sorted naïve and memory T cells showed that alloreactive precursors were equally present in both populations. The CD8+ effector and terminally differentiated effector memory subsets contained the highest proportion of granzyme B/perforin after allostimulation, suggesting that these cells present a significant threat to transplanted organs. Finally, we demonstrate that virus-specific lymphocytes contribute significantly to the alloresponse in certain responder-stimulator HLA combinations, underscoring the importance of T-cell cross-reactivity in alloimmunity. These results provide a quantitative assessment of the roles of naïve and memory T-cell subsets in the normal human alloimmune response and establish a platform for measuring T-cell alloreactivity pre- and posttransplantation.


Subject(s)
T-Lymphocytes/cytology , Adult , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Female , Flow Cytometry/methods , Granzymes/pharmacology , HLA Antigens/chemistry , Humans , Immunologic Memory/immunology , Interferon-gamma/metabolism , Male , Perforin/metabolism , T-Lymphocyte Subsets/immunology
11.
Am J Transplant ; 7(4): 949-54, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17331114

ABSTRACT

Lymphoid clusters (LC) containing CD20-positive B cells in kidney allografts undergoing acute cellular rejection (ACR) have been identified in small studies as a prognostic factor for glucocorticoid resistance and graft loss. Allograft biopsies obtained during the first episode of ACR in 120 recipients were evaluated for LC, immunostained with CD20 antibody, and correlated with conventional histopathologic criteria, response to treatment and outcome. LC were found in 71 (59%) of the 120 biopsies. All contained CD20 positive B cells that accounted for 5-90% of the LC leukocyte content. The incidence of LC was highest in the patients who had no lymphoid depletion or had been treated with Thymoglobulin preconditioning (79% vs. 75%, respectively) compared to 37% in patients pretreated with Campath (p = 0.0001). Banff 1a/1b ACR were more frequent in the LC-positive than the LC-negative group (96% vs. 80%, respectively; p = 0.0051). With a posttransplant follow-up of 953 +/- 430 days, no significant differences were detected between LC-postitive and LC-negative groups in time to ACR, steroid resistance, serum creatinine and graft loss. CD20+LC did not portend glucocorticoid resistance or worse short to medium term outcomes. CD20+LC may represent a heterogenous collection in which there may be a small still to be fully defined unfavorable subgroup.


Subject(s)
Antigens, CD20/immunology , B-Lymphocytes/pathology , Graft Rejection/drug therapy , Graft Rejection/immunology , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/immunology , Lymphocyte Depletion/adverse effects , Acute Disease , Adult , Alemtuzumab , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/therapeutic use , Antigens, CD/immunology , Antilymphocyte Serum/therapeutic use , B-Lymphocytes/immunology , Biopsy , Female , Graft Rejection/pathology , Humans , Kidney Transplantation/pathology , Male , Middle Aged , Retrospective Studies
12.
Am J Transplant ; 7(5): 1071-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17359505

ABSTRACT

Tertiary lymphoid tissues are lymph node-like cell aggregates that arise at sites of chronic inflammation. They have been observed in transplanted organs undergoing chronic rejection, but it is not known whether they contribute to the rejection process by supporting local activation of naïve lymphocytes. To answer this question, we established a murine transplantation model in which the donor skin contains tertiary lymphoid tissues due to transgenic expression of lymphotoxin-alpha(RIP-LT alpha), whereas the recipient lacks all secondary lymphoid organs and does not mount primary alloimmune responses. We demonstrate in this model that RIP-LT alpha allografts that harbor tertiary lymphoid tissues are rejected, while wild-type allografts that lack tertiary lymphoid tissues are accepted. Wild-type allografts transplanted at the same time as RIP-LT alpha skin or 60 days later were also rejected, suggesting that tertiary lymphoid tissues, similar to secondary lymphoid organs, generate both effector and memory immune responses. Consistent with this observation, naive T cells transferred to RIP-LT alpha skin allograft but not syngeneic graft recipients proliferated and differentiated into effector and memory T cells. These findings provide direct evidence that tertiary lymphoid structures perpetuate the rejection process by supporting naïve T-cell activation.


Subject(s)
Graft Rejection/immunology , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Skin Transplantation/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cell Proliferation , Female , Graft Rejection/pathology , Lymphoid Tissue/pathology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Skin/immunology , Skin/metabolism , Skin/pathology , Skin Transplantation/pathology , T-Lymphocytes/pathology , Transplantation, Homologous
13.
J Immunol ; 167(12): 6711-5, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739484

ABSTRACT

Despite declining thymic output with age, the peripheral naive T cell pool of an adult animal remains remarkably stable. Therefore, a central question in immunology is how the naive T cell pool is maintained. Here we show that the maintenance of the naive CD4, but not CD8, T cell population in the thymectomized adult mouse is dependent on the presence of secondary lymphoid tissues. This finding is explained by the inability of naive CD4 T cells to sustain normal levels of the survival molecule Bcl-2 or to undergo homeostatic proliferation in the absence of secondary lymphoid organs. Thus, naive CD4 T cells must traffic through secondary lymphoid organs to maintain a stable CD4 pool while naive CD8 T cells encounter their survival and proliferation signals outside the organized structures of secondary lymphoid tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphoid Tissue/immunology , Animals , Cells, Cultured , Homeostasis , Hyaluronan Receptors/analysis , Immunophenotyping , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Peyer's Patches/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Spleen/immunology , Splenectomy , T-Lymphocyte Subsets/classification , Thymectomy
14.
Transplantation ; 72(5): 948-53, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11571464

ABSTRACT

BACKGROUND: High-density oligoarray technology is a novel method for screening the expression of thousands of genes in a small tissue sample. Oligoarray analysis of genes expressed during human renal allograft rejection has not been reported previously. METHODS: Seven human renal allograft biopsies with histologic evidence of acute cellular rejection and three renal allograft biopsies without evidence of rejection (control) were analyzed for the expression of 6800 human genes using high-density oligoarrays (GeneChip, Affymetrix, Santa Clara, CA). Quantitative expression of gene transcripts was determined and a comparison analysis between acute rejection and control biopsy samples was performed. Up-regulation of a specific gene transcript during acute rejection was considered to be significant if transcript abundance increased fourfold or more relative to control biopsy samples. RESULTS: Comparison analysis revealed that between 32 and 219 gene transcripts are up-regulated (>fourfold) during acute rejection. Of these transcripts, only four (human monokine induced by interferon-gamma, T-cell receptor active beta-chain protein, interleukin-2 stimulated phosphoprotein, and RING4 (a transporter involved in antigen presentation)) were consistently up-regulated in each acute rejection sample relative to at least two of three control biopsy samples. Six other genes were up-regulated in six of seven acute rejection samples. These were interferon-stimulated growth factor-3, complement factor 3, nicotinamide N-methyltransferase, macrophage inflammatory protein-3beta, myeloid differentiation protein, and CD18. Only two gene transcripts were down-regulated in five of seven acute rejection samples. Significant up-regulation of cytotoxic T-cell effector molecules, previously reported as markers of acute renal rejection in humans, was not detected. CONCLUSIONS: High-density oligoarray technology is useful for screening gene expression in transplanted tissues undergoing acute rejection. Because this method does not rely on a priori knowledge of which genes are involved in acute rejection, it is likely to yield novel insights into the mechanisms and diagnosis of rejection.


Subject(s)
Gene Expression , Intercellular Signaling Peptides and Proteins , Kidney Transplantation/immunology , Oligonucleotide Array Sequence Analysis , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , Adult , Case-Control Studies , Chemokine CCL19 , Chemokine CXCL9 , Chemokines, CC/genetics , Chemokines, CXC/genetics , Female , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Kidney Transplantation/physiology , Male , Middle Aged , Phosphoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology
15.
J Immunol ; 167(2): 1125-8, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11441125

ABSTRACT

Current evidence suggests that neonatal tolerance to a foreign Ag is the consequence of IL-4-mediated Th2 immunity rather than the thymic deletion of Ag-specific T cells. Here, we addressed the role of IL-4 in neonatal tolerance by testing whether tolerance to a minor histocompatibility Ag can be induced in newborn mice that lack IL-4 (IL-4(-/-)). We found that IL-4 does not play a dominant role in the induction of neonatal tolerance as newborn female IL-4(-/-) mice could be readily tolerized to the H-Y male Ag. In contrast, mice that lack both IL-4 and IL-13 (IL-4(-/-)/IL-13(-/-)) were resistant to the induction of neonatal tolerance, and their splenocytes produced exaggerated amounts of IFN-gamma on rechallenge with the same Ag encountered during the neonatal period. These findings argue against the view that IL-4 alone is critical for the induction of neonatal tolerance and suggest that the combined actions of both IL-4 and IL-13 are essential for this process.


Subject(s)
Animals, Newborn/genetics , Animals, Newborn/immunology , Immune Tolerance/genetics , Interleukin-13/deficiency , Interleukin-13/genetics , Interleukin-4/deficiency , Interleukin-4/genetics , Animals , Female , H-Y Antigen/administration & dosage , H-Y Antigen/immunology , Injections, Intraperitoneal , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-4/physiology , Lymphocyte Transfusion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Skin Transplantation/immunology , Spleen/cytology , Spleen/transplantation , Th2 Cells/immunology
17.
J Immunol ; 165(6): 3031-6, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975812

ABSTRACT

The mechanisms responsible for the generation and maintenance of T cell memory are unclear. In this study, we tested the role of IL-2 in allospecific CD8+ T cell memory by analyzing the long-term survival, phenotype, and functional characteristics of IL-2-replete (IL-2+/+) and IL-2-deficient (IL-2-/-) CD8+ TCR-transgenic lymphocytes in an adoptive transfer model. We found that IL-2 is not essential for the in vivo generation, maintenance, or recall response of CD8+ memory T cells. However, IL-2 increased the size of the CD8+ memory pool if present at the time of initial T cell activation but reduced the size of the pool if present during memory maintenance by inhibiting the proliferation of CD8+ memory T cells. Thus, IL-2-based vaccine strategies or immunosuppressive regimens that target IL-2 should take into account the divergent roles of IL-2 in CD8+ T cell immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-2/physiology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Survival/immunology , Epitopes, T-Lymphocyte/immunology , Immunophenotyping , Injections, Intravenous , Interleukin-2/deficiency , Interleukin-2/genetics , Interleukin-2/pharmacology , Lymphocyte Count , Lymphocyte Transfusion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/transplantation
18.
Nat Med ; 6(6): 686-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835686

ABSTRACT

Secondary lymphoid organs (the spleen, lymph nodes and mucosal lymphoid tissues) provide the proper environment for antigen-presenting cells to interact with and activate naive T and B lymphocytes. Although it is generally accepted that secondary lymphoid organs are essential for initiating immune responses to microbial antigens and to skin allografts, the prevailing view has been that the immune response to primarily vascularized organ transplants such as hearts and kidneys does not require the presence of secondary lymphoid tissue. The assumption has been that the immune response to such organs is initiated in the graft itself when recipient lymphocytes encounter foreign histocompatibility antigens presented by the graft's endothelial cells. In contrast to this view, we show here that cardiac allografts are accepted indefinitely in recipient mice that lack secondary lymphoid tissue, indicating that the alloimmune response to a vascularized organ transplant cannot be initiated in the graft itself. Moreover, we demonstrate that the permanent acceptance of these grafts is not due to tolerance but is because of immunologic 'ignorance'.


Subject(s)
Heart Transplantation/immunology , Lymphoid Tissue/immunology , Animals , Graft Rejection , Immune Tolerance/immunology , Isoantigens/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/immunology , Skin Transplantation/immunology , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
19.
Transplantation ; 69(11): 2428-32, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10868653

ABSTRACT

BACKGROUND: Fas ligand (FasL)-Fas and tumor necrosis factor alpha (TNFalpha)-tumor necrosis factor receptor (TNFR) interactions regulate immune responses and contribute to self-tolerance by mediating antigen-driven T cell apoptosis. It is not known whether FasL and TNFalpha, expressed by the recipient's lymphoid or nonlymphoid cells, are essential for the apoptosis of alloreactive T lymphocytes and the induction of allograft acceptance. METHODS: We compared the survival of fully allogeneic vascularized cardiac allografts between wild-type (wt) and FasL-mutant (gld) recipient mice. In addition, we studied cardiac allograft survival in gld mice injected with TNFalpha-neutralizing antibody. Allograft acceptance (graft survival >100 days) was induced by treating the recipients with CTLA4Ig, a recombinant fusion protein that blocks B7-CD28 T cell costimulation. In vivo alloantigen-driven apoptosis of mature CD4+ and CD8+ T lymphocytes was analyzed in mice repeatedly stimulated with allogeneic splenocytes. RESULTS: We found that CTLA4Ig induces 100% long-term acceptance of cardiac allografts in wt and gld mice. Similarly, CTLA4Ig induced 100% allograft acceptance in gld recipients injected with TNFalpha-neutralizing antibody. In vivo alloantigen-driven apoptosis of mature CD4+ and CD8+ T cells was significantly reduced in gld mice and in wt mice treated with anti-TNFalpha antibody. However, neutralizing TNFalpha activity in gld mice failed to abrogate alloantigen-driven T cell apoptosis. CONCLUSIONS: These data indicate that: (1) FasL and TNFalpha expression are not obligatory for the induction of long-term allograft acceptance by CTLA4Ig and (2) FasL- and TNFalpha-independent death pathways contribute to alloantigen-driven T cell apoptosis.


Subject(s)
Graft Survival/physiology , Isoantigens/physiology , Membrane Glycoproteins/physiology , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/physiology , Acute Disease , Animals , Apoptosis/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Death/physiology , Fas Ligand Protein , Graft Rejection/etiology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Transplantation, Homologous
20.
Semin Nephrol ; 20(2): 95-102, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10746853

ABSTRACT

Allograft rejection is the consequence of the recipient's alloimmune response to nonself antigens expressed by donor tissues. Allospecific T lymphocytes are activated by donor peptides which are presented in the context of major histocompatibility complex molecules by either donor or recipient antigen presenting cells. Antigen presenting cells also provide essential costimulatory signals which are required for T cell proliferation and differentiation into either helper or effector lymphocytes. Effector mechanisms of allograft rejection include those mediated by cytotoxic T lymphocytes, macrophages, natural killer cells, and B lymphocytes. Importantly, alloimmune responses are controlled by regulatory molecules which include membrane receptors and cytokines. Novel insights into the interactions between antigen presenting cells and T lymphocytes, and further understanding of how alloimmune responses are regulated, will help in developing effective antirejection and tolerance-inducing strategies.


Subject(s)
Graft Rejection/immunology , Antigen-Presenting Cells/immunology , Cytokines/immunology , Humans , Isoantibodies/immunology , Isoantigens/immunology , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...