Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
2.
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
3.
Am J Transplant ; 15(5): 1231-40, 2015 May.
Article in English | MEDLINE | ID: mdl-25676865

ABSTRACT

Vascularized composite allotransplantation (VCA) has emerged as a treatment option for treating nonlife-threatening conditions. Therefore, in order to make VCA a safe reconstruction option, there is a need to minimize immunosuppression, develop tolerance-inducing strategies and elucidate the mechanisms of VCA rejection and tolerance. In this study we explored the effects of hIL-2/Fc (a long-lasting human IL-2 fusion protein), in combination with antilymphocyte serum (ALS) and short-term cyclosporine A (CsA), on graft survival, regulatory T cell (Treg) proliferation and tolerance induction in a rat hind-limb transplant model. We demonstrate that hIL-2/Fc therapy tips the immune balance, increasing Treg proliferation and suppressing effector T cells, and permits VCA tolerance as demonstrated by long-term allograft survival and donor-antigen acceptance. Moreover, we observe two distinct types of acute rejection (AR), progressive and reversible, within hIL-2/Fc plus ALS and CsA treated recipients. Our study shows differential gene expression profiles of FoxP3 versus GzmB, Prf1 or interferon-γ in these two types of AR, with reversible rejection demonstrating higher Treg to Teff gene expression. This correlation of gene expression profile at the first clinical sign of AR with VCA outcomes can provide the basis for further inquiry into the mechanistic aspects of VCA rejection and future drug targets.


Subject(s)
Hindlimb/transplantation , Interleukin-2/chemistry , Recombinant Fusion Proteins/chemistry , Transplantation Tolerance/drug effects , Vascularized Composite Allotransplantation/methods , Animals , Cell Proliferation , Cyclosporine/chemistry , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Graft Rejection , Graft Survival , Granzymes/metabolism , Humans , Immune System , Immune Tolerance , Male , Pore Forming Cytotoxic Proteins/metabolism , Rats , Rats, Inbred Lew , Rats, Wistar , Transplantation, Homologous
4.
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
5.
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
6.
Am J Transplant ; 13(8): 2161-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23718897

ABSTRACT

Vascularized composite allografts (VCAs) are unique among transplanted organs in that they are composed of multiple tissues with disparate antigenic and immunologic properties. As the predominant indications for VCAs are non-life-threatening conditions, there is an immediate need to develop tolerance induction strategies and to elucidate the mechanisms of VCA rejection and tolerance using VCA-specific animal models. In this study, we explore the effects of in vitro induced donor antigen-specific CD4(-) CD8(-) double negative (DN) Treg-based therapy, in a fully MHC mismatched mouse VCA such as a vascularized osteomyocutaneous as compared to a non-VCA such as a full thickness skin (FTS) transplantation model to elucidate the unique features of VCA rejection and tolerance. We demonstrate that combined therapy with antigen-induced CD4 derived DN Tregs and a short course of anti-lymphocyte serum, rapamycin and IL-2/Fc fusion protein results in donor-specific tolerance to VCA, but not FTS allografts. Macrochimerism was detected in VCA but not FTS allograft recipients up to >60 days after transplantation. Moreover, a significant increase of CD4(+) Foxp3(+) Tregs was found in the peripheral blood of tolerant VCA recipients. These data suggest that VCA are permissive to tolerance induced by DN Treg-based induction therapy.


Subject(s)
Bone Transplantation/immunology , Graft Survival/immunology , Immunomodulation , Muscle, Skeletal/transplantation , Skin Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance , Animals , CD4 Antigens/metabolism , Cell Proliferation , Flow Cytometry , Graft Rejection/immunology , Interleukin-2/pharmacology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Transplantation Chimera , Transplantation Conditioning , Transplantation, Homologous
7.
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
8.
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
9.
Diabetologia ; 54(8): 2082-92, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21594554

ABSTRACT

AIMS/HYPOTHESIS: A new differentiation pathway for CD4(-)CD8(-) (DN) T cells has recently been identified that exhibits the potent function of peripheral converted DN T cells in suppressing immune responses and provides the potential to treat autoimmune diseases. The aim of this study was to determine if the DN T cells converted from CD4(+) T cells of NOD mice retain the antigen-specific regulatory capacity and prevent autoimmune diabetes in vivo. We also sought to determine if the combination of DN T cells with rapamycin promotes islet allograft survival in autoimmune diabetic NOD recipients. METHODS: NOD CD4(+) T cells were converted to DN T cells in an in vitro mixed-lymphocyte reaction, with or without GAD65 peptide, as previously reported. The antigen-specific DN T cells were adoptively transferred to NOD/SCID mice, new-onset diabetic NOD mice or islet-allograft-recipient NOD mice as the part of cell-based therapy. The development of diabetes and allograft survival was assessed by monitoring blood glucose levels. RESULTS: NOD CD4(+) T cells were converted in vitro to DN T cells at a rate of 50% and expressed unique cell features. The DN T cells from NOD donors blocked autoimmunity and prevented diabetes in NOD models, and these effects were even greater for GAD65-peptide-primed DN T cells. DN T cells acted in conjunction with rapamycin to suppress alloantigen-triggered T cell proliferation, promoted apoptosis and prolonged islet allograft survival in NOD recipients. CONCLUSIONS/INTERPRETATION: Administration of the islet beta cell antigen-specific DN T cells can prevent the development of autoimmune diabetes and promote islet allograft survival in NOD mice.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus, Type 1/prevention & control , Islets of Langerhans Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Female , Flow Cytometry , Male , Mice , Mice, Inbred NOD , Polymerase Chain Reaction
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Br J Ophthalmol ; 93(9): 1134-40, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19286686

ABSTRACT

Maintenance of ocular viability is one of the major impediments to successful whole-eye transplantation. This review provides a comprehensive understanding of the current literature to help guide future studies in order to overcome this hurdle. A systematic multistage review of published literature was performed. Three specific questions were addressed: (1) Is recovery of visual function following eye transplantation greater in cold-blooded vertebrates when compared with mammals? (2) Is outer retina function following enucleation and reperfusion improved compared with enucleation alone? (3) Following optic-nerve transection, is there a correlation between retinal ganglion cell (RGC) survival and either time after transection or proximity of the transection to the globe? In a majority of the studies performed in the literature, recovery of visual function can occur after whole-eye transplantation in cold-blooded vertebrates. Following enucleation (and reperfusion), outer retinal function is maintained from 4 to 9 h. RGC survival following optic-nerve transection is inversely related to both the time since transection and the proximity of transection to the globe. Lastly, neurotrophins can increase RGC survival following optic-nerve transection. This review of the literature suggests that the use of a donor eye is feasible for whole-eye transplantation.


Subject(s)
Eye Enucleation/adverse effects , Eye/transplantation , Optic Nerve Injuries/complications , Retinal Ganglion Cells/physiology , Animals , Eye/physiopathology , Optic Nerve Injuries/physiopathology , Retina/physiology , Tissue Donors , Tissue Survival/physiology , Visual Acuity
16.
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
17.
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
18.
Transplant Proc ; 36(2 Suppl): 495S-499S, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15041395

ABSTRACT

Everolimus is a derivative of sirolimus, a macrocyclic lactone, originally isolated from Streptomyces hygroscopicus. Both everolimus and sirolimus have a similar mechanism of action, exerting potent inhibition of growth factor-induced proliferation of lymphocytes, as well as other hematopoietic and nonhematopoietic cells of mesenchymal origin. Each agent complexes with the FK506 binding protein 12 to inhibit cyclin dependent kinase(s), collectively termed the target of rapamycin (TOR), causing G1-S phase cell cycle arrest. Safety and efficacy have been documented in large-scale, blinded, randomized, international clinical renal and cardiac transplant trials. Everolimus is more hydrophilic, exhibits a shorter elimination half-life (approximately 30 hours), and demonstrates greater relative bioavailability compared to sirolimus. However, similar to the calcineurin inhibitors and sirolimus, everolimus is biotransformed by the cytochrome P450, 3A4 isozyme. Also similar to sirolimus, clinical experiences identified biologically relevant side effects including hyperlipidemia and exacerbation of cyclosporine (CsA)-associated nephrotoxicity. However, also similar to sirolimus, accumulating evidence suggests that the hyperlipidemia can be controlled and the CsA-associated renal effects appear reduced with a low incidence of acute rejection when everolimus is administered in combination with reduced CsA doses. The experience using everolimus in cardiac transplantation has also provided potentially important insights into the consequences of antiproliferative effects on vascular smooth muscle cells and fibroblasts where reduction in intimal expansion was identified by intravascular coronary ultrasound examination among those patients receiving everolimus. Therefore, available results suggest that the introduction of everolimus as the newest TOR inhibitor should enhance therapeutic options for immunosuppression after organ transplantation.


Subject(s)
Immunosuppressive Agents/therapeutic use , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use , Transplantation Immunology , Clinical Trials, Phase I as Topic , Everolimus , Humans , Immunosuppressive Agents/toxicity , Sirolimus/toxicity
19.
Transplant Proc ; 35(3 Suppl): 95S-98S, 2003 May.
Article in English | MEDLINE | ID: mdl-12742475

ABSTRACT

Sirolimus (SRL), a fermentation product of Streptomyces hygroscopicus, complexes with the FKBP12 to inhibit cyclin dependent kinase(s), collectively termed the target of rapamycin (TOR), causing G(1)-S phase cell cycle arrest. Safety and efficacy have been documented in clinical renal transplantation, but concerns were raised due to important biologically relevant side effects. Hyperlipidemia was identified, beginning with early clinical experiences, and the unexpected findings that SRL may exacerbate CsA associated nephrotoxicity was observed during the pivotal phase III studies. This report details results of our experience using SRL (target trough concentration, 10-15 ng/mL) with low dose CsA (target trough concentration, 50-100 ng/mL), seeking to determine whether this approach might provide effective immunosuppression while reducing associated nephrotoxicity. Among 121 renal transplant recipients, 62 received the SRL based regimen and 59 received MMF with all patients receiving CsA and prednisone. Similar to earlier clinical experiences, hematopoeitic abnormalities and hyperlipidemia were observed among patients who received SRL, and those abnormalities were readily controlled. However, unlike observations from the phase III SRL studies, renal function was not adversely affected. These findings support the growing body of evidence indicating that SRL based immunosuppression in combination low dose calcineurin inhibitors and corticosteroids is safe, efficacious, and without associated renal toxicity.


Subject(s)
Cyclosporine/therapeutic use , Kidney Transplantation/physiology , Sirolimus/therapeutic use , Tacrolimus/therapeutic use , Blood Pressure , Creatinine/blood , Cyclosporine/administration & dosage , Dose-Response Relationship, Drug , Follow-Up Studies , Graft Rejection/epidemiology , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/immunology , Postoperative Complications/epidemiology , Reoperation/statistics & numerical data , Tacrolimus/administration & dosage , Tacrolimus Binding Proteins/metabolism , Time Factors
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...