Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Heart Lung Transplant ; 24(11): 1906-14, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16297799

ABSTRACT

BACKGROUND: Prolonged cold ischemia is thought to exacerbate ischemia-reperfusion injury and graft coronary artery disease (GCAD). We investigated the effect of varying lengths of cold ischemia on inflammation and apoptosis during ischemia-reperfusion injury and correlated this with the degree of GCAD in rat cardiac allografts. METHODS: PVG rat (RT1c) hearts subjected to 30, 60, 90, 120, or 150 minutes of cold ischemia were heterotopically transplanted into ACI rats (RT1a). Grafts were procured after 4 hours of reperfusion and analyzed for superoxide generation, myeloperoxidase activity, tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and monocyte chemoattractant protein-1/chemokine (C-C motif) ligand 2 (MCP-1/CCL2) production, cardiomyocyte apoptosis, and caspase-2, -3, -8, -9 activities. Additional transplanted animals received cyclosporine A (7.5 mg/kg/day) for 10 days as chronic rejection models. Indices of GCAD were determined at 90 days. RESULTS: A direct linear correlation was found between cold ischemic time, ischemia-reperfusion injury, and GCAD. Superoxide generation, myeloperoxidase activity, TNF-alpha, IL-1beta, MCP-1/CCL2 production, cardiomyocyte apoptosis, and caspase-2, -3, -8, and -9 activities increased with ischemic time, peaking at 120 minutes and plateauing at 150 minutes. GCAD, assessed by the percentage of luminal narrowing, the intima/media ratio, and the percentage of diseased vessels, worsened with increased ischemic time, peaking at 120 minutes and plateauing at 150 minutes. All tested variables in both the acute and chronic phases were significantly increased with 120-minute ischemia compared with 30-minute ischemia. CONCLUSIONS: These data indicate that the degree of cardiomyocyte apoptosis and inflammatory response in cardiac allografts during ischemia-reperfusion injury depends on the duration of cold ischemia. More important, that prolonged cold ischemia correlates with increased GCAD.


Subject(s)
Cold Ischemia/adverse effects , Coronary Disease/etiology , Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/pathology , Animals , Apoptosis , Caspases/metabolism , Chemokine CCL2/metabolism , Enzyme-Linked Immunosorbent Assay , Heart Transplantation/immunology , In Situ Nick-End Labeling , Interleukin-1/metabolism , Male , Myocardial Reperfusion Injury/etiology , Myocytes, Cardiac , Rats , Rats, Inbred ACI , Rats, Inbred Strains , Superoxide Dismutase/metabolism , Time Factors , Transplantation, Heterotopic , Tumor Necrosis Factor-alpha/metabolism
2.
Am J Transplant ; 5(6): 1286-96, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15888032

ABSTRACT

Chronic rejection remains the major obstacle for long-term transplant survival. Both indirect alloresponse and tissue-specific autoimmunity have been implicated in its pathogenesis. The interrelationship between these two types of host anti-graft response remains poorly understood. We have developed an immunosuppression-free mouse model of graft coronary artery disease (GCAD), in which all FVB (H-2(q)) cardiac allografts placed into minor Ag (mHC)-mismatched DBA/1 (H-2(q)) hosts survived more than 112 days, and developed GCAD. We then examined the kinetics of both anti-mHC alloresponse and host autoimmunity against heart-specific antigen, cardiac myosin (CM). At 8 days post-transplantation, recipient mice showed minimal intragraft inflammation and apoptosis, and limited expansion of allo-specific T cells. In addition, we observed early production of anti-myosin IgG1 autoantibodies, which occurred in the absence of activated CM-specific T lymphocytes. By day 56, GCAD indices, the numbers of mHC- and CM-reactive T cells, and the levels of circulating allo- and CM-specific antibodies were all significantly increased. While host alloresponse was exhausted at 112 days post-transplant, T-cell reactivity against CM persisted. Our data suggest that both allo- and tissue-specific immunity might contribute to the induction of GCAD. They indicate that continual autoimmune response against graft tissue antigens may provide for GCAD sustenance.


Subject(s)
Autoimmunity/immunology , Cardiac Myosins/immunology , Coronary Artery Disease/immunology , Graft Rejection/immunology , Graft Survival , Heart Transplantation/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation , Cardiac Myosins/metabolism , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Progression , Histocompatibility Antigens Class I/immunology , Immune Tolerance , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , T-Lymphocytes/metabolism , Transplantation, Homologous
3.
J Heart Lung Transplant ; 24(4): 446-53, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15797747

ABSTRACT

BACKGROUND: The effect of cyclosporine (CsA) on the development of graft coronary artery disease (GCAD) is controversial. We developed a novel allogeneic mouse model of heart transplantation and investigated the effect of CsA on acute rejection and GCAD. METHODS: Hearts of FVB mice (H-2(q)) were heterotopically transplanted into 60 C57BL/6 mice (H-2(b)). CsA was administered to recipients at 10, 20 or 30 mg/kg/day for 10 or 30 days after transplantation. Untreated recipients as well as isograft recipients served as controls. Viability of the grafts was assessed daily by palpation. Parenchymal rejection was scored in grafts surviving 30 days in the 30-day treatment groups. GCAD was evaluated by the percentage of luminal narrowing, intima/media ratio and percentage of diseased vessels. Blood CsA and creatinine levels were also evaluated. Results were evaluated statistically. RESULTS: All groups except the untreated control group and the allograft groups treated with 10 or 20 mg for 10 days showed significant graft survival (>/=33% survival for 30 days). An inverse correlation was observed between CsA treatment dose, parenchymal rejection score and degree of GCAD in the 30-day treatment groups. However, graft survival in the 20-mg/kg/day group was significantly better than that in the 30-mg/kg/day group. Serum creatine levels showed no nephrotoxicity. CONCLUSIONS: Relatively high-dose CsA mitigated parenchymal rejection and GCAD of the mouse cardiac allografts. In addition, a valuable mouse model mimicking the clinical course of GCAD was achieved with CsA treatment of 20 mg/kg/day for 30 days.


Subject(s)
Coronary Artery Disease/drug therapy , Cyclosporine/therapeutic use , Disease Models, Animal , Heart Transplantation , Immunosuppressive Agents/therapeutic use , Animals , Chromatography, Liquid , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Coronary Vessels/pathology , Cyclosporine/administration & dosage , Cyclosporine/pharmacokinetics , Dose-Response Relationship, Drug , Follow-Up Studies , Gas Chromatography-Mass Spectrometry , Graft Rejection/blood , Graft Rejection/pathology , Graft Rejection/prevention & control , Graft Survival , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Monocytes/pathology , Myocytes, Cardiac/pathology , Transplantation, Homologous , Treatment Outcome
4.
Circulation ; 110(11 Suppl 1): II200-6, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364863

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury is an important risk factor for graft coronary artery disease (GCAD). We hypothesized that overexpression of SOD1 in donor hearts would suppress ischemia-reperfusion injury and thereby reduce GCAD. METHODS AND RESULTS: In one series, donor hearts of C57BL/6 (H-2b) transgenic mice overexpressing human SOD1 or C57BL/6 wild-type mice were heterotopically transplanted into C57BL/6 recipients and procured after 4 hours of reperfusion (n=6 each). Superoxide, TNF-alpha, and MCP-1/CCL2 production were significantly reduced in the SOD1 transgenic donor heart recipients, and graft injury determined by serum CPK-MB levels was significantly decreased. Cardiomyocyte apoptosis and caspase-3 and caspase-9 activities were significantly decreased in these recipients; caspase-8 activity was unchanged. Fas ligand but not Fas expression was also reduced. In a second series, transgenic and wild-type hearts were transplanted into C-H-2bm12KhEg (H-2bm12) recipients, and then procured on day 56 (n=7 each). Cardiac graft beating was significantly better in the SOD1 transgenic donor heart recipients on days 28, 42, and 56 (but not day 14). Significant reduction in luminal narrowing, the intima/media ratio, and the percentage of diseased vessels was seen in the SOD1 transgenic donor heart recipients, and MCP-1/CCL2, ICAM-1, and VCAM-1 production were significantly reduced. CONCLUSIONS: Overexpression of SOD1 attenuates both apoptosis and the inflammatory response during ischemia-reperfusion injury and therefore mitigates against the subsequent development of GCAD.


Subject(s)
Coronary Disease/prevention & control , Graft Rejection/prevention & control , Heart Transplantation , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Postoperative Complications/prevention & control , Superoxide Dismutase/physiology , Animals , Apoptosis , Caspase 3 , Caspase 9 , Caspases/metabolism , Chemokine CCL2/biosynthesis , Coronary Disease/etiology , Drug Evaluation, Preclinical , Fas Ligand Protein , Graft Rejection/etiology , Humans , Inflammation/prevention & control , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Contraction , Myocardial Ischemia/complications , Myocardial Reperfusion Injury/complications , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Peroxidase/analysis , Postoperative Complications/etiology , Recombinant Fusion Proteins/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Superoxides/metabolism , Transplantation, Heterotopic , Transplantation, Homologous , Tumor Necrosis Factor-alpha/biosynthesis
5.
Circulation ; 110(11 Suppl 1): II194-9, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364862

ABSTRACT

BACKGROUND: Inhibiting delta protein kinase C (deltaPKC) during reperfusion and activating epsilon PKC (epsilonPKC) before ischemia each limits cardiac ischemic injury. Here, we examined whether limiting ischemia-reperfusion injury inhibits graft coronary artery disease (GCAD) and improves murine cardiac allografting. METHODS AND RESULTS: Hearts of FVB mice (H-2q) were transplanted into C57BL/6 mice (H-2b). epsilonPKC activator (psiepsilonRACK) was injected intraperitoneally (20 nmol) into donor mice 20 minutes before procurement. Hearts were then perfused with psiepsilonRACK (1.5 nmol) through the inferior vena cava (IVC) and subsequently submerged in psiepsilonRACK (0.5 micromol/L) for 20 minutes at 4 degrees C. Before reperfusion, the peritoneal cavity of recipients was irrigated with deltaPKC inhibitor (deltaV1-1, 300 nmol); control animals were treated with normal saline. The total ischemic time to the organ was 50 minutes. Two hours after transplantation, production of inflammatory cytokines and adhesion molecules, cardiomyocyte apoptosis, and caspase-3 and caspase-9 (but not caspase-8) activities were significantly reduced in the PKC regulator-treated group. Fas ligand levels (but not Fas) were also significantly reduced in this group. Importantly, GCAD indices, production of inflammatory cytokines, and adhesion molecules were significantly decreased and cardiac allograft function was significantly better as measured up to 30 days after transplantation. CONCLUSIONS: An epsilonPKC activator and a deltaPKC inhibitor together reduced GCAD. Clinically, these PKC isozyme regulators may be useful for organ preservation and prevention of ischemia-reperfusion injury and graft coronary artery disease in cardiac transplantation.


Subject(s)
Coronary Disease/drug therapy , Graft Rejection/drug therapy , Heart Transplantation , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Oligopeptides/therapeutic use , Postoperative Complications/drug therapy , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Animals , Apoptosis/drug effects , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Adhesion Molecules/biosynthesis , Coronary Disease/etiology , Cytokines/biosynthesis , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Fas Ligand Protein , Graft Rejection/etiology , Inflammation/prevention & control , Injections, Intraperitoneal , Male , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Myocardial Ischemia/complications , Myocardial Reperfusion Injury/complications , Myocytes, Cardiac/pathology , Necrosis , Oligopeptides/administration & dosage , Peroxidase/analysis , Postoperative Complications/etiology , Protein Kinase C-delta , Protein Kinase C-epsilon , Therapeutic Irrigation , Transplantation, Heterotopic , Transplantation, Homologous
6.
Blood ; 104(12): 3789-96, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15280201

ABSTRACT

After cardiac transplantation, graft damage occurs secondary to ischemia-reperfusion injury and acute rejection. This damage ultimately leads to the development of graft coronary artery disease (GCAD), which limits long-term graft survival. Apoptosis is directly involved in graft injury, contributing to the development of GCAD. To assess the role of the antiapoptotic factor Bcl-2 in the process of GCAD, we transplanted hearts from FVB transgenic mice overexpressing human Bcl-2 under the control of alpha-myosin heavy chain promoter into allogenic C57BL/6 mice. Bcl-2 overexpression led to reduced cytochrome c-mediated caspase-9-dependent cardiomyocyte apoptosis and local inflammation (neutrophil infiltration and proinflammatory cytokine production) in cardiac allografts during ischemia-reperfusion injury and also led to reduced immune responses (inflammatory cell infiltration, production of T(H)1 cytokines and chemokines, and expression of adhesion molecules) during acute and chronic rejection without affecting host CD4(+) and CD8(+) cell responses in the spleen. Thus, local Bcl-2 expression directly contributes to the modulation of local immune responses in allograft rejection, resulting in attenuated GCAD. In conclusion, our findings suggest that the modulation of Bcl-2 expression by pharmacologic up-regulation or gene transfer may be of clinical benefit in the short- and long-term function of cardiac allografts.


Subject(s)
Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Reperfusion Injury/therapy , Animals , Apoptosis/drug effects , Coronary Artery Disease/etiology , Coronary Artery Disease/prevention & control , Coronary Artery Disease/therapy , Genetic Vectors , Graft Rejection/immunology , Humans , Immunity/drug effects , Inflammation/drug therapy , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/administration & dosage , Proto-Oncogene Proteins c-bcl-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...