Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
2.
J Endocrinol ; 218(1): 75-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23589741

ABSTRACT

Islets are exceptionally susceptible to ischemia-reperfusion injury, an increased incidence of primary graft nonfunctionality, and ß-cell death during a transplant procedure. Therefore, islets require protection during the early stages of the transplant procedure. Based on the beneficial vascular and anti-inflammatory activity of adiponectin, we hypothesize that adiponectin protects islet cells against ischemia-reperfusion injury and graft dysfunction after transplantation. To examine the effects of adiponectin on the resistance of islet ischemia-reperfusion injury, we used the islet hypoxia-reoxygenation injury model and performed kidney subcapsular syngeneic islet transplants to assess the islets' vitality and function. Furthermore, we utilized lipopolysaccharide (LPS)-induced or tumor necrosis factor α (TNFα)-induced damage to islet cells to model the inflammation of post-transplant ischemia-reperfusion injury and transplanted islets in adiponectin knockout mice to explore whether the protective action of adiponectin is involved in TNFα production and nuclear transcription factor-κB (NF-κB) activation. Adiponectin suppressed TNFα production and IκB-α phosphorylation; decreased hypoxia-reoxygenation and LPS-induced and TNFα-induced islet apoptosis; and improved islet function in vivo and in vitro. Our results demonstrate that adiponectin protects the islet from injury. We show that islet protection occurs in response to ischemia-reperfusion and is dependent on the suppression of islet production by TNFα through cyclooxygenase 2 and the inhibition of the TNFα-induced NF-κB activation pathways.


Subject(s)
Adiponectin/metabolism , Cyclooxygenase 2/metabolism , Islets of Langerhans/metabolism , NF-kappa B/metabolism , Reperfusion Injury/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Adiponectin/antagonists & inhibitors , Adiponectin/genetics , Adiponectin/pharmacology , Animals , Apoptosis/drug effects , Cell Hypoxia , Hormone Antagonists/pharmacology , I-kappa B Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Male , Mice , Mice, Knockout , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Protective Agents/chemistry , Protective Agents/metabolism , Protective Agents/pharmacology , Protein Processing, Post-Translational/drug effects , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Tissue Culture Techniques , Transplantation, Heterotopic/immunology , Tumor Necrosis Factor-alpha/pharmacology
3.
Diabetes ; 62(5): 1646-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23423564

ABSTRACT

Transplantation of adult pancreatic islets has been proposed to cure type 1 diabetes (T1D). However, it is rarely considered in the clinic because of its transient effect on disease, the paucity of donors, and the requirement for strong immunosuppressive treatment to prevent allogeneic graft rejection. Transplantation of fetal pancreases (FPs) may constitute an attractive alternative because of potential abundant donor sources, possible long-term effects due to the presence of stem cells maintaining tissue integrity, and their supposed low immunogenicity. In this work, we studied the capacity of early FPs from mouse embryos to develop into functional pancreatic islets producing insulin after transplantation in syngeneic and allogeneic recipients. We found that as few as two FPs were sufficient to control T1D in syngeneic mice. Surprisingly, their development into insulin-producing cells was significantly delayed in male compared with female recipients, which may be explained by lower levels of prolactin in males. Finally, allogeneic FPs were rapidly rejected, even in the context of minor histocompatibility disparities, with massive graft infiltration with T and myeloid cells. This work suggests that FP transplantation as a therapeutic option of T1D needs to be further assessed and would require immunosuppressive treatment.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Embryo, Mammalian , Fetus , Pancreas Transplantation/methods , Prolactin/therapeutic use , Transplantation, Heterotopic/methods , Animals , Cell Differentiation , Crosses, Genetic , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , Islets of Langerhans Transplantation/pathology , Kidney , Male , Mice , Mice, Knockout , Mice, Nude , Mice, Transgenic , Pancreas Transplantation/immunology , Pancreas Transplantation/pathology , Sex Characteristics , Specific Pathogen-Free Organisms , Transplantation, Heterotopic/immunology , Transplantation, Heterotopic/pathology , Transplantation, Homologous , Transplantation, Isogeneic
4.
Diabetes ; 62(5): 1665-75, 2013 May.
Article in English | MEDLINE | ID: mdl-23315496

ABSTRACT

The lymphocytic ionotropic purinergic P2X receptors (P2X1R-P2X7R, or P2XRs) sense ATP released during cell damage-activation, thus regulating T-cell activation. We aim to define the role of P2XRs during islet allograft rejection and to establish a novel anti-P2XRs strategy to achieve long-term islet allograft function. Our data demonstrate that P2X1R and P2X7R are induced in islet allograft-infiltrating cells, that only P2X7R is increasingly expressed during alloimmune response, and that P2X1R is augmented in both allogeneic and syngeneic transplantation. In vivo short-term P2X7R targeting (using periodate-oxidized ATP [oATP]) delays islet allograft rejection, reduces the frequency of Th1/Th17 cells, and induces hyporesponsiveness toward donor antigens. oATP-treated mice displayed preserved islet grafts with reduced Th1 transcripts. P2X7R targeting and rapamycin synergized in inducing long-term islet function in 80% of transplanted mice and resulted in reshaping of the recipient immune system. In vitro P2X7R targeting using oATP reduced T-cell activation and diminished Th1/Th17 cytokine production. Peripheral blood mononuclear cells obtained from long-term islet-transplanted patients showed an increased percentage of P2X7R⁺CD4⁺ T cells compared with controls. The beneficial effects of oATP treatment revealed a role for the purinergic system in islet allograft rejection, and the targeting of P2X7R is a novel strategy to induce long-term islet allograft function.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Graft Rejection/prevention & control , Immunosuppression Therapy , Islets of Langerhans Transplantation/adverse effects , Purinergic P2X Receptor Antagonists/therapeutic use , Receptors, Purinergic P2X7/metabolism , Transplantation, Heterotopic/adverse effects , Adenosine Triphosphate/therapeutic use , Adult , Animals , Female , Humans , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics , Sirolimus/therapeutic use , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Transplantation, Heterotopic/immunology , Transplantation, Heterotopic/pathology , Transplantation, Homologous , Transplantation, Isogeneic
5.
Diabetes ; 62(3): 905-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23160528

ABSTRACT

It is generally believed that inflammatory cues can attract noncognate, "bystander" T-cell specificities to sites of inflammation. We have shown that recruitment of naive and in vitro activated autoreactive CD8⁺ T cells into endogenous islets requires local autoantigen expression. Here, we demonstrate that absence of an autoantigen in syngeneic extrapancreatic islet grafts in diabetic hosts renders the grafts "invisible" to cognate memory (and naive) T cells. We monitored the recruitment of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)206₋214-reactive CD8⁺ T cells into IGRP206₋214-competent and IGRP206₋214-deficient islet grafts in diabetic wild-type or IGRP206₋214(-/-) nonobese diabetic hosts (harboring either naive and memory T cells or only naive IGRP206₋214-specific T-cells, respectively). All four host-donor combinations had development of recurrent diabetes within 2 weeks. Wild-type hosts recruited IGRP206₋214-specific T cells into IGRP206₋214(+/+) but not IGRP206₋214(-/-) grafts. In IGRP206₋214(-/-) hosts, there was no recruitment of IGRP206₋214-specific T cells, regardless of donor type. Graft-derived IGRP206₋214 activated naive IGRP206₋214-specific T cells, but graft destruction invariably predated their recruitment. These results indicate that recurrent diabetes is exclusively driven by autoreactive T cells primed during the primary autoimmune response, and demonstrate that local antigen expression is a sine qua non requirement for accumulation of memory T cells into islet grafts. These findings underscore the importance of tackling autoreactive T-cell memory after ß-cell replacement therapy.


Subject(s)
Autoantigens/analysis , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/therapy , Immunologic Memory , Islets of Langerhans Transplantation/immunology , Transplantation, Heterotopic/immunology , Animals , Autoantigens/genetics , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Epitopes , Glucose-6-Phosphatase/analysis , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Graft Survival , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans Transplantation/pathology , Kidney , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred NOD , Mice, Transgenic , Pancreas/immunology , Pancreas/metabolism , Pancreas/pathology , Peptide Fragments/analysis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteins/analysis , Proteins/genetics , Proteins/metabolism , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Transplantation, Heterotopic/adverse effects , Transplantation, Isogeneic/adverse effects , Transplantation, Isogeneic/immunology
6.
Acta Cardiol ; 66(6): 779-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22299390

ABSTRACT

OBJECTIVE: Acute cardiac allograft rejection continues to be the cause of graft loss and contributes to the morbidity and mortality after cardiac transplantation. Repetitive endomyocardial biopsies are necessary to monitor the effects of immunosuppressants after cardiac transplantation. In this study, we investigate whether speckle tracking imaging (STI) is a valuable method in assessing acute cardiac rejection. METHODS AND RESULTS: Hearts from Brown Norway rats or Lewis rats were transplanted into other Brown Norway rats. Isografts and groups of allografts, either untreated or treated with cyclosporine A (CsA) at a low dose (3 mg x kg(-1) x d(-1)) or a high dose (10 mg x kg(-1) x d(-1)), were compared 7 days after transplantation. Echocardiography-derived left ventricular post wall thickness was increased only in untreated allografts.The left ventricular ejection fraction was significantly lower in the allografts compared with the isografts, but allografts treated without or with low-dose CsA showed similar results. The radial velocity and systolic radial strain rate showed a lower value in untreated allografts than other grafts, but there is no significant difference between allografts treated with high- or low-dose CsA and isografts. The circumferential strain and circumferential strain rate were comparable among the 4 groups. However, the radial strain exhibited a clear gradient in these groups (2.8 +/- 1.3 in untreated allografts, 5.2 +/- 10.9 in allografts treated with low-dose CsA, 6.3 +/- 1.8 in allografts treated with high-dose CsA, and 12.7 +/- 7.9 in isografts, P < 0.001). CONCLUSIONS: STI is able to offer a noninvasive method for detecting transplant allograft rejection.


Subject(s)
Echocardiography/methods , Graft Rejection/diagnostic imaging , Heart Transplantation/immunology , Animals , Graft Rejection/pathology , Heart Transplantation/methods , Myocardium/pathology , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Heterotopic/immunology
7.
Transplant Proc ; 42(6): 2149-51, 2010.
Article in English | MEDLINE | ID: mdl-20692430

ABSTRACT

A modified immunosuppressive regimen, developed at the National Institutes of Health, has been employed in a large animal model of heterotopic cardiac xenotransplantation. Graft survival has been prolonged, but despite this, our recipients have succumbed to various surgical or nonsurgical complications. Herein, we have described different complications and management strategies. The most common complication was hypercoagulability (HC) after transplantation, causing thrombosis of both small and large vasculature, ultimately leading to graft loss. While managing this complication we discovered that there was a delicate balance between HC and consumptive coagulopathy (CC). CC encountered in some recipient baboons was not able to be reversed by stopping anticoagulation and administering multiple blood transfusions. Some complications had iatrogenic components. To monitor the animals, a solid state left ventricular telemetry probe was placed directly into the transplanted heart via the apex. Induction of hypocoagulable states by continuous heparin infusion led to uncontrollable intra-abdominal bleeding in 1 baboon from this apical site. This occurrence necessitated securing the probe more tightly with multiple purse strings and 4-quadrant pledgeted stay sutures. One instance of cardiac rupture originated from a lateral wall infarction site. Earlier studies have shown infections to be uniformly fatal in this transplant model. However, owing to the telemetry placement, infections were identified early by temperature spikes that were treated promptly with antibiotics. We had several cases of wound dehiscence due to recipients disrupting the suture line. These complications were promptly resolved by either re-approximating the wound or finding distractions for the baboon. A few of the most common problems we faced in our earlier experiments were related to the jacket, tether, and infusion pumps. It was difficult to keep the jackets on some baboons and the tether had to be modified several times before we assured long-term success. Infusion catheter replacement resulted in transplant heart venous obstruction and thrombosis from a right common femoral venous line. Homeostatic perturbations such as HC and CC and baboon-induced wound complications comprised most complications. Major bleeding and death due to telemetry implantation and infarct rupture occurred in 2 baboons. Despite the variety of complications, we achieved significant graft prolongation in this model.


Subject(s)
Heart Transplantation/adverse effects , Intraoperative Complications/classification , Postoperative Complications/classification , Transplantation, Heterologous/adverse effects , Transplantation, Heterotopic/adverse effects , Anastomosis, Surgical/methods , Animals , Aorta, Abdominal/surgery , Immunosuppressive Agents/therapeutic use , Papio , Pulmonary Artery/surgery , Swine , Transplantation, Heterologous/immunology , Transplantation, Heterotopic/immunology , Vena Cava, Inferior/surgery
8.
Transplantation ; 90(1): 4-13, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20463646

ABSTRACT

BACKGROUND: There is increasing evidence for a role for autoimmunity in transplant rejection. It has previously been shown that autoantibodies to vimentin (Vim) accelerate acute rejection of murine cardiac allografts. We have investigated whether autoimmunity to Vim contributes to development of cardiac allograft vasculopathy (CAV). METHODS: Two well-established minor mismatch murine models of CAV were used, transplantation of 129/sv hearts into T-cell-depleted C57Bl/6 (B6) recipients and transplantation of FVB hearts into nonimmunosuppressed DBA/1 recipients. Recipients were immunized with recombinant mouse Vim in complete Freunds adjuvant, and controls received hen egg lysozyme 2 weeks before transplantation. T cell and antibody responses to Vim were assessed by ELISPOT and ELISA, respectively. CAV within transplanted hearts was assessed by quantitative morphometry of occluded vessels, presence of smooth muscle cells, deposition of C3d, and confocal microscopy. RESULTS: Allografts were harvested from B6 recipients at days 30 and 45 and from DBA/1 recipients at days 18 and 35. At all days, there was significantly more intimal occlusion of arteries of Vim -immunized mice than controls. There was significantly more smooth muscle cell alpha actin in vessels from Vim-immunized mice, and more C3d deposited in hearts from Vim-immunized mice. Confocal microscopy demonstrated colocalization of Vim with C3d on endothelial cells, leukocytes, and platelets in allogeneic but not syngeneic hearts. Serum from Vim-immunized mice, but not controls, caused platelet/leukocyte conjugation when added to mouse leukocytes. CONCLUSION: The autoimmune response to Vim accelerates CAV progression in these minor-mismatched models.


Subject(s)
Heart Transplantation/immunology , Transplantation, Homologous/immunology , Transplantation, Isogeneic/immunology , Vimentin/immunology , Animals , Antibodies, Monoclonal/pharmacology , Autoimmunity/immunology , Enzyme-Linked Immunosorbent Assay , Graft Rejection/pathology , Heart Transplantation/pathology , Histocompatibility Testing , Leukocytes/drug effects , Leukocytes/immunology , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microscopy, Confocal , Minor Histocompatibility Antigens/immunology , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Transplantation, Heterotopic/immunology
9.
Transplant Proc ; 42(3): 976-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20430219

ABSTRACT

Inhibition of T-cell activation is the most efficient way to prevent transplant rejection. Protein kinase C (PKC) is an important signaling enzyme in the activation and regulation of T lymphocytes. AEB-071 (AEB) is a low-molecular-weight compound that blocks early T-cell activation via selective inhibition of PKC, a mechanism that differs from that of the calcineurin inhibitors. The present study sought to compare the effects of AEB versus tacrolimus (Tac) to prevent acute rejection in rats that had undergone heterotopic heart transplantation. We investigated the Brown Norway-Lewis rat strain combination for cardiac graft survival over 30 days after transplantation using varying doses of oral AEB and Tac monotherapy. Grafts were monitored by daily palpation; cessation of palpable ventricular contraction was considered to be rejection. Apart from necropsy, we performed histologic examinations of cardiac graft at 7 days after transplantation. In untreated recipients, allograft mean survival times (MST) was 6.83+/-0.41 days. AEB at 15, 30, or 60 mg/kg versus Tac at 1.2 mg/kg significantly prolonged graft survival to a MST of 12.33+/-1.21, 16.67+/-1.21, and 19.33+/-3.83, versus 17.00+/-6.90 days, respectively. Histologic assessment at 7 days after transplantation showed that high-dose AEB significantly decreased the histologic rejection score, indicative of decreased inflammatory cell infiltration into the graft. These results suggested that the administration of AEB (medium or high-dose), a PKC inhibitor, mitigated acute rejection and displayed significantly longer MST, similar to high-dose Tac after heterotopic heart transplantation in the rat.


Subject(s)
Graft Rejection/prevention & control , Heart Transplantation/immunology , Pyrroles/therapeutic use , Quinazolines/therapeutic use , Tacrolimus/therapeutic use , Animals , Graft Survival/drug effects , Heart Transplantation/physiology , Lymphocyte Activation/drug effects , Male , Myocardial Contraction , Rats , Rats, Inbred BN , Rats, Inbred Lew , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Heterotopic/immunology , Transplantation, Homologous
10.
Transplantation ; 88(4): 465-9, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19696628

ABSTRACT

BACKGROUND: The aim of our study was to examine the feasibility of allogeneic uterine transplantation in a large animal model. METHODS: We performed heterotopic uterine transplants in genetically defined mini-pigs. Immunosuppression was tacrolimus administered intravenously for the first 12 days posttransplantation followed by oral cyclosporine maintenance immunosuppression. The graft was transplanted heterotopically in the lower abdominal cavity of the recipient. The vaginal vault was exteriorized as a stoma in the lower right abdominal wall. The uterine grafts were followed with endoscopies and biopsies. RESULTS: Ten transplants were performed. Follow-up was until July 2008. At the end of the follow-up period, 5 animals were alive and healthy, 0.5 to 12 months posttransplantation. There were 5 deaths due to pneumonia (n=1), intussusception of the graft (n=1), cardiorespiratory arrest during anesthesia (n=1), and complications of the stoma (n=2). Acute rejections of the graft presented during the 2nd and 3rd month posttransplantation were treated successfully with increase of the maintenance immunosuppression and steroids. Other complications included prolapse and infections of the graft stoma. Pathological changes seen in the endometrial biopsies included acute rejection and acute endometritis. CONCLUSION: These findings demonstrate that successful uterus transplantation in a large animal model (miniature swine) is feasible using this heterotopic model, and it can be useful for the study of these transplants.


Subject(s)
Uterus/transplantation , Animals , Endometritis/etiology , Endometritis/pathology , Female , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival , Models, Animal , Swine , Swine, Miniature , Transplantation, Heterotopic/adverse effects , Transplantation, Heterotopic/immunology , Transplantation, Heterotopic/methods , Transplantation, Homologous , Uterus/immunology , Uterus/pathology
11.
J Heart Lung Transplant ; 28(2): 176-82, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19201344

ABSTRACT

BACKGROUND: It has been suggested that pregnancy-related hormones play a critical role in mediating selective immune tolerance during pregnancy. An understanding of why a woman's body normally does not reject the fetus is highly relevant to the prevention of transplant rejection. METHODS: The hearts of female inbred F344 rats (RT-1(lvl)) were transplanted into naive Lewis (RT-1(l); nLewis) or pregnant (pLewis; Day 6, 12 and 18 of pregnancy) rats. The mean survival time (MST) of the cardiac allografts between the nLewis and pLewis rats was compared. We determined the rate of proliferation of the T cells isolated from nLewis and pLewis rats in response to concanavalin A (ConA), anti-CD3 and -CD28 antibody and alloantigen stimulation ex vivo. mRNA expression of several cytokines in these T cells was analyzed using quantitative reverse transcriptase-polymerase chain reaction (RT-PCR). In addition, the effect of estriol on the cardiac allograft was tested. RESULTS: The pLewis rats with transplant on Day 12 of pregnancy had the most significantly prolonged F344 cardiac graft survival (MST 13.3 days) as compared with nLewis recipients (MST 8 days). pLewis T-cell proliferation was stimulated by alloantigen and antibody but ConA was reduced, whereas Th1/Th2 cytokine mRNA profiles in the T cells were similar for nLewis and pLewis rats. Likewise, estriol also significantly prolonged survival of cardiac allografts. CONCLUSIONS: The results of this study demonstrate that pregnancy hormones not only appear to play a critical role in maternal acceptance of the fetus, but also have therapeutic potential for prolonging the survival of major histocompatibility complex (MHC)-compatible allografts during pregnancy.


Subject(s)
Heart Transplantation/immunology , Major Histocompatibility Complex , Pregnancy, Animal/immunology , Animals , Concanavalin A/pharmacology , Cytokines/genetics , Estriol/therapeutic use , Female , Graft Survival/drug effects , Heart Transplantation/pathology , Lymphocyte Activation/drug effects , Male , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Heterotopic/immunology , Transplantation, Heterotopic/pathology , Transplantation, Homologous/pathology
12.
Biol Pharm Bull ; 31(2): 305-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18239292

ABSTRACT

Histone deacetylase (HDAC) inhibitors repress interleukin-2 (IL-2) gene expression in T cells and possess immunosuppressive activity in vivo. In addition to its immunosuppressive activity, HDAC inhibitors block GATA binding protein-1 (GATA-1) gene expression in megakaryocytes and elicit thrombocytopenia. In this report we state that for a given immunosuppressive dose of HDAC inhibitor, the ratio of GATA-1 reporter gene activity relative to IL-2 reporter gene assay (G/I ratio of measured IC(50)) can be predictive of a HDAC inhibitor's thrombocytopenic effect. This study utilized nine HDAC inhibitors at a minimal effective dose in a rat heterotopic cardiac transplantation model and the resultant G/I ratios and platelet depletion rates were highly correlated (r=0.933). These results indicate that calculation of G/I ratio can be a novel method for selecting immunosuppressive HDAC inhibitor having minimal thrombocytopenic effect which will benefit the search for new immunosuppressants of greater safety and efficacy.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Immunosuppressive Agents/pharmacology , Thrombocytopenia/chemically induced , Animals , Cell Survival/drug effects , Drug Evaluation, Preclinical , Enzyme Inhibitors/adverse effects , GATA1 Transcription Factor/genetics , Genes, Reporter/drug effects , Heart Transplantation/immunology , Humans , Immunosuppressive Agents/adverse effects , Interleukin-2/genetics , Jurkat Cells , Male , Plasmids/genetics , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Tetrazolium Salts , Thiazoles , Thrombocytopenia/blood , Transplantation, Heterotopic/immunology
13.
Blood ; 110(10): 3691-4, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17684157

ABSTRACT

Mesenchymal stem cells (MSCs) display immunomodulatory properties mediated by various factors, including inducible nitric oxide synthase (iNOS). Since heme oxygenase-1 (HO-1) is a potent immunosuppressive enzyme, we tested the hypothesis that HO-1 could mediate the immunosuppressive effects of MSCs. We generated adult rat MSCs that inhibited T-cell proliferation in vitro. These MSCs expressed both HO-1 and iNOS. In vitro, whereas neither HO-1 nor iNOS inhibition alone could interfere with the immunosuppressive properties of rat MSCs, simultaneous inhibition of both enzymes restored T-cell proliferation. In vivo, injection of MSCs significantly delayed heart allograft rejection, and inhibition of either HO-1 or iNOS totally reversed the protective activity of MSCs, inducing rejection. Adult human MSCs also expressed HO-1; in these cells, HO-1 inhibition was sufficient to completely block their immunosuppressive capacity. In conclusion, we show, for the first time, that HO-1 mediates the immunosuppressive properties of rat and human MSCs.


Subject(s)
Heme Oxygenase-1/physiology , Immune Tolerance/genetics , Mesenchymal Stem Cells/physiology , Animals , Cells, Cultured , Graft Survival , Heart Transplantation/immunology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Metalloporphyrins/pharmacology , Protoporphyrins/pharmacology , Rats , Transplantation Tolerance/immunology , Transplantation, Heterotopic/immunology
14.
Transplantation ; 83(6): 815-8, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17414717

ABSTRACT

We developed a mouse model of heterotopic limb transplantation in which we took advantage of Thy1.1 and Thy1.2 congenic strains to track and characterize donor T cells, to determine the role of recipient's thymus in mixed T-cell chimerism induction as well as transplant immunocompetence. The vascularized Thy1.1 limb graft composed of femur, muscle, and skin (VBT) survived long-term in more than 87.5% of Thy1.2 recipients. Percentages of donor-type Thy1.1 T cells increased from day 30 to 90 in thymus and spleen of recipients. Most peripheral donor T cells displayed a naïve phenotype and a few others were regulatory T cells. Thymectomy prevented peripheral T-cell chimerism. Congenic VBT in immunodeficient RAG mice restored their ability to reject skin allografts. These observations suggest that donor T cells differentiated in host thymus might contribute to maintenance of mixed chimerism after transplantation of tissue composite grafts that include vascularized bone.


Subject(s)
Extremities/transplantation , T-Lymphocytes/cytology , Thymus Gland/cytology , Transplantation, Heterotopic/immunology , Animals , Bone Marrow Transplantation/immunology , Cell Proliferation , Graft Rejection/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/immunology , Thy-1 Antigens/metabolism , Thymus Gland/immunology , Transplantation Chimera/immunology
15.
Transpl Int ; 20(3): 256-65, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17291219

ABSTRACT

Correlative data suggest that mast cells adversely affect cardiac transplantation. This study uses a mast cell-deficient rat model to directly address the role of mast cells in cardiac allotransplantation. Standardized cardiac heterotopic transplantation with cyclosporine immunosuppression was performed in mast cell-deficient and mast cell-competent rats. Rejection, ischemia, fibrosis, fibrin deposition, numbers of T-cell receptor alpha/beta positive cells, expression of transforming growth factor-beta (TGF-beta), and of endothelin-1 (ET-1) and its receptors ETA and ETB were assessed. Differences in baseline cardiac gene expression were quantified by real-time PCR in a separate group of untransplanted animals. Baseline cardiac gene expression levels of all investigated growth factors, cytokines, ET-1, ETA, and ETB were similar in mast cell-deficient and mast cell-competent rats. Surprisingly, upon heterotopic transplantation, donor heart survival was significantly reduced in mast cell-deficient rats. Moreover, in mast cell-deficient donor hearts rejection was more severe, although nonsignificant, and extracellular matrix associated TGF-beta immunoreactivity was significantly lower than in mast cell-competent donor hearts. Fibrin immunoreactive area, on the other hand, was only increased in mast cell-deficient donor hearts, but not in mast cell-competent donor hearts. Histopathological changes in all donor hearts were accompanied by increased immunoreactivity for ET-1. In conclusion, this study shows that mast cells play a protective role after cardiac transplantation.


Subject(s)
Heart Transplantation/immunology , Mast Cells/physiology , Animals , Cytokines/biosynthesis , Graft Survival/physiology , Male , Models, Animal , Rats , Transplantation, Heterotopic/immunology
16.
Transplant Proc ; 38(6): 1853-4, 2006.
Article in English | MEDLINE | ID: mdl-16908304

ABSTRACT

BACKGROUND: The amount of native small bowel required for adequate nutrition is variable, but lies between 10% and 20% of full length. Currently, for patients requiring small bowel transplantation (SBT), standard practice is to transplant the entire small bowel if space permits. Few experimental studies have addressed the effect of the length of small bowel transplanted on immune responses and in those that have, the amount of mesenteric lymph node (MLN) transplanted has always been a potential confounding factor, as have differences between jejunum and ileum. METHODS: Full-length and segmental heterotopic rat SBT was performed between PVG donor and DA recipients. To transplant reduced length small bowel grafts but to exclude immunologic differences between jejunum and ileum, equal lengths of bowel were resected from proximal and distal ends in the donor. A proportional amount of MLN was carefully dissected using a microvascular technique and then excised. Serial serum samples from the transplant recipients were tested for anti-PVG (rejection) and anti-DA (graft-versus-host) antibodies using a two-color flow cytometric technique, described previously, with the aim of looking for differences in immunologic responses to full and segmental grafts. RESULTS: We have established a model of segmental SBT that includes a proportional amount of MLN and is free from differences between jejunum and ileum. Preliminary data have demonstrated the development of circulating anti-host and anti-graft antibodies with time for both full-length and segmental SBT.


Subject(s)
Intestine, Small/transplantation , Transplantation, Homologous/immunology , Animals , Isoantibodies/blood , Models, Animal , Rats , Rats, Inbred Strains , Transplantation, Heterotopic/immunology
17.
PLoS Med ; 3(7): e215, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16768546

ABSTRACT

BACKGROUND: Transplantation of embryonic pig pancreatic tissue as a source of insulin has been suggested for the cure of diabetes. However, previous limited clinical trials failed in their attempts to treat diabetic patients by transplantation of advanced gestational age porcine embryonic pancreas. In the present study we examined growth potential, functionality, and immunogenicity of pig embryonic pancreatic tissue harvested at different gestational ages. METHODS AND FINDINGS: Implantation of embryonic pig pancreatic tissues of different gestational ages in SCID mice reveals that embryonic day 42 (E42) pig pancreas can enable a massive growth of pig islets for prolonged periods and restore normoglycemia in diabetic mice. Furthermore, both direct and indirect T cell rejection responses to the xenogeneic tissue demonstrated that E42 tissue, in comparison to E56 or later embryonic tissues, exhibits markedly reduced immunogenicity. Finally, fully immunocompetent diabetic mice grafted with the E42 pig pancreatic tissue and treated with an immunosuppression protocol comprising CTLA4-Ig and anti-CD40 ligand (anti-CD40L) attained normal blood glucose levels, eliminating the need for insulin. CONCLUSIONS: These results emphasize the importance of selecting embryonic tissue of the correct gestational age for optimal growth and function and for reduced immunogenicity, and provide a proof of principle for the therapeutic potential of E42 embryonic pig pancreatic tissue transplantation in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Pancreas Transplantation , Transplantation, Heterologous , Abatacept , Agammaglobulinaemia Tyrosine Kinase , Alloxan , Animals , Blood Glucose/analysis , CD40 Ligand/antagonists & inhibitors , Female , Gestational Age , Graft Rejection/prevention & control , Humans , Immunocompetence , Immunoconjugates/therapeutic use , Immunosuppressive Agents/therapeutic use , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Kidney , Leukocytes, Mononuclear/transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Mice, Mutant Strains , Mice, Nude , Mice, SCID , Pancreas/embryology , Pancreas Transplantation/immunology , Pancreas, Exocrine/ultrastructure , Pregnancy , Protein-Tyrosine Kinases/deficiency , Sus scrofa/embryology , Transplantation, Heterologous/immunology , Transplantation, Heterotopic/immunology
18.
Eur Surg Res ; 37(4): 220-7, 2005.
Article in English | MEDLINE | ID: mdl-16260872

ABSTRACT

OBJECTIVE: The aim of this study was to assess the effect of immunoregulatory cytokine interleukin-10 (IL-10) gene therapy on pancreas tissue rejection in a heterotopic pancreas transplantation model. BACKGROUND: Modulation of inflammatory responses by anti-inflammatory cytokines (e.g., IL-10) has been suggested to minimize organ rejection. In this context, modulation of cytokines using gene therapy could be a new therapeutic modality in preventing organ rejection. METHODS: The study was performed using male inbred Wistar rats as recipients and Sprague-Dawley rats as donors. 24 h before transplantation, groups of rats, named IL-10 (n = 20) and green fluorescent protein (GFP, n = 20), were injected with viral vectors Ad5CMVhIL10 or Ad5CMVGFP. Sham-operated rats (n = 20) underwent saline injection only before transplantation. The pancreatic tissue from each of these donor rats was subsequently transplanted into the corresponding groups of streptozotocin-induced diabetic recipient rats. Recipients were thus transfected with either IL-10 (n = 20), GFP-only carrying viral vectors (n = 20) or no viral vectors (normal saline, n = 20). A selected number of animals from each recipient group (n = 5) was sacrificed at weekly intervals for 3 weeks and some were further followed up to 12 weeks before sacrifice. Histological assessment of the pancreatic tissue was made based on rejection and GFP expression. Blood glucose levels were checked daily in all groups until sacrifice. Upon sacrifice, serum cytokine and insulin levels were measured. Histopathological correlations between blood glucose levels, serum insulin levels and serum IL-10 levels were made. RESULTS: IL-10 gene therapy significantly attenuated pancreas rejection compared to controls, provided more normal blood glucose levels and elevated plasma insulin levels. Upon assumed natural deactivation of transferred viruses after 4 weeks, differences between groups in terms of rejection, blood glucose and insulin levels disappeared. CONCLUSION: These findings suggest that IL-10 gene therapy significantly reduced pancreas rejection.


Subject(s)
Genetic Therapy , Graft Rejection/immunology , Interleukin-10/genetics , Pancreas Transplantation/immunology , Transfection , Transplantation, Heterotopic/immunology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Insulin/metabolism , Interleukin-10/metabolism , Male , Pancreas/cytology , Pancreas/pathology , Pancreas/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Rats, Wistar
19.
BMC Immunol ; 6: 22, 2005 Sep 21.
Article in English | MEDLINE | ID: mdl-16176581

ABSTRACT

BACKGROUND: Immune activation that results due to the aberrant proliferation of lymphocytes leads to inflammation and graft rejection in organ transplant recipients. We hypothesize that the cell cycle control and inflammation are parallel events, inhibition of cellular proliferation by cyclin kinase inhibitor specifically p21 will limit inflammation and prevent allograft rejection. METHODS: We performed in vitro and in vivo studies using lymphocytes, and rat heart transplant model to understand the role of cyclins and p21 on mitogen and allo-induced lymphocyte activation and inflammation. Lymphocyte proliferation was studied by 3H-thymidine uptake assay and mRNA expression was studied RT-PCR. RESULTS: Activation of allo- and mitogen stimulated lymphocytes resulted in increased expression of cyclins, IL-2 and pro-inflammatory cytokines, which was inhibited by cyclosporine. The over-expression of p21 prolonged graft survival in a completely mismatched rat heart transplant model resulted by inhibiting circulating and intra-graft expression of proinflammatory cytokines. CONCLUSION: Cyclins play a significant role in transplant-induced immune activation and p21 over-expression has potential to inhibit T cell activation and inflammation. The results from this study will permit the design of alternate strategies by controlling cell cycle progression to achieve immunosuppression in transplantation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclins/physiology , Cytokines/physiology , Graft Rejection/physiopathology , Inflammation/physiopathology , Lymphocyte Activation , Animals , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclosporine/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Profiling , Graft Rejection/immunology , Heart Transplantation/immunology , Humans , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Interleukin-2/biosynthesis , Interleukin-2/genetics , Jurkat Cells/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Lymphocytes/drug effects , Lymphocytes/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Rats, Inbred WF , Recombinant Fusion Proteins/physiology , Transplantation, Heterotopic/immunology , Transplantation, Homologous/immunology
20.
Circulation ; 112(9 Suppl): I166-72, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16159810

ABSTRACT

BACKGROUND: We investigated whether differentiation of embryonic stem cells (ESCs) in ischemic myocardium enhances their immunogenicity, thereby increasing their chance for rejection. METHODS AND RESULTS: In one series, 129/SvJ-derived mouse ESCs (ES-D3 line) were transplanted by direct myocardial injection (1 x 10(6) cells) into murine hearts of both allogeneic (BALB/c, n=20) and syngeneic (129/SvJ, n=12) recipients after left anterior artery ligation. Hearts were procured at 1, 2, 4, and 8 weeks after ESC transplantation and analyzed by immunohistochemistry to assess immune cell infiltration (CD3, CD4, CD8, B220, CD11c, Mac-1, and Gr-1) and ESC differentiation (hematoxylin and eosin). In a second series (allogeneic n=5, sham n=3), ESC transplantation was performed similarly; however after 2 weeks, left anterior descending artery-ligated and ESC-injected hearts were heterotopically transplanted into naive BALB/c recipients. After an additional 2 weeks, donor hearts were procured and analyzed by immunohistochemistry. In the first series, the size of all ESC grafts remained stable and there was no evidence of ESC differentiation 2 weeks after transplantation; however, after 4 weeks, both allogeneic and syngeneic ESC grafts showed the presence of teratoma. By 8 weeks, surviving ESCs could be detected in the syngeneic but not in the allogeneic group. Mild inflammatory cellular infiltrates were found in allogeneic recipients at 1 and 2 weeks after transplantation, progressing into vigorous infiltration at 4 and 8 weeks. The second series demonstrated similar vigorous infiltration of immune cells as early as 2 weeks after heterotopic transplantation. CONCLUSIONS: In vivo differentiated ESCs elicit an accelerated immune response as compared with undifferentiated ESCs. These data imply that clinical transplantation of allogeneic ESCs or ESC derivatives for treatment of cardiac failure might require immunosuppressive therapy.


Subject(s)
Cell Differentiation/immunology , Fetal Tissue Transplantation/immunology , Graft Rejection/immunology , Heart Neoplasms/etiology , Myocardial Ischemia/surgery , Myocardium/immunology , Pluripotent Stem Cells/transplantation , Postoperative Complications/etiology , Stem Cell Transplantation , Teratoma/etiology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line/immunology , Cell Line/transplantation , Dendritic Cells/immunology , Female , Granulocytes/immunology , Heart Neoplasms/pathology , Heart Transplantation/immunology , Injections, Intramuscular , Macrophages/immunology , Mice , Mice, Inbred BALB C , Myocardium/pathology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/immunology , Postoperative Complications/pathology , Stem Cell Transplantation/adverse effects , T-Lymphocyte Subsets/immunology , Teratoma/pathology , Transplantation, Heterotopic/immunology , Transplantation, Homologous/immunology , Transplantation, Isogeneic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...