Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Transplant ; 16(4): 1160-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26689983

RESUMO

Organ damage and innate immunity during heart transplantation may evoke adaptive immunity with serious consequences. Because lymphatic vessels bridge innate and adaptive immunity, they are critical in immune surveillance; however, their role in ischemia-reperfusion injury (IRI) in allotransplantation remains unknown. We investigated whether the lymphangiogenic VEGF-C/VEGFR3 pathway during cardiac allograft IRI regulates organ damage and subsequent interplay between innate and adaptive immunity. We found that cardiac allograft IRI, within hours, increased graft VEGF-C expression and lymphatic vessel activation in the form of increased lymphatic VEGFR3 and adhesion protein expression. Pharmacological VEGF-C/VEGFR3 stimulation resulted in early lymphatic activation and later increase in allograft inflammation. In contrast, pharmacological VEGF-C/VEGFR3 inhibition during cardiac allograft IRI decreased early lymphatic vessel activation with subsequent dampening of acute and chronic rejection. Genetic deletion of VEGFR3 specifically in the lymphatics of the transplanted heart recapitulated the survival effect achieved by pharmacological VEGF-C/VEGFR3 inhibition. Our results suggest that tissue damage rapidly changes lymphatic vessel phenotype, which, in turn, may shape the interplay of innate and adaptive immunity. Importantly, VEGF-C/VEGFR3 inhibition during solid organ transplant IRI could be used as lymphatic-targeted immunomodulatory therapy to prevent acute and chronic rejection.


Assuntos
Células Endoteliais/metabolismo , Rejeição de Enxerto/metabolismo , Transplante de Coração , Traumatismo por Reperfusão/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Aloenxertos , Animais , Células Endoteliais/patologia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/imunologia , Inflamação/metabolismo , Inflamação/patologia , Linfangiogênese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Traumatismo por Reperfusão/fisiopatologia , Doadores de Tecidos , Fator C de Crescimento do Endotélio Vascular/metabolismo
2.
Am J Transplant ; 15(8): 2075-84, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25932532

RESUMO

The major cause of death during the first year after heart transplantation is primary graft dysfunction due to preservation and ischemia-reperfusion injury (IRI). Angiopoietin-1 is a Tie2 receptor-binding paracrine growth factor with anti-inflammatory properties and indispensable roles in vascular development and stability. We used a stable variant of angiopoietin-1 (COMP-Ang1) to test whether ex vivo intracoronary treatment with a single dose of COMP-Ang1 in donor Dark Agouti rat heart subjected to 4-h cold ischemia would prevent microvascular dysfunction and inflammatory responses in the fully allogeneic recipient Wistar Furth rat. COMP-Ang1 reduced endothelial cell-cell junction disruption of the donor heart in transmission electron microscopy during 4-h cold ischemia, improved myocardial reflow, and reduced microvascular leakage and cardiomyocyte injury of transplanted allografts during IRI. Concurrently, the treatment reduced expression of danger signals, dendritic cell maturation markers, endothelial cell adhesion molecule VCAM-1 and RhoA/Rho-associated protein kinase activation and the influx of macrophages and neutrophils. Furthermore, COMP-Ang1 treatment provided sustained anti-inflammatory effects during acute rejection and prevented the development of cardiac fibrosis and allograft vasculopathy. These results suggest donor heart treatment with COMP-Ang1 having important clinical implications in the prevention of primary and subsequent long-term injury and dysfunction in cardiac allografts.


Assuntos
Rejeição de Enxerto/prevenção & controle , Transplante de Coração , Proteínas Recombinantes de Fusão/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Doadores de Tecidos , Animais , Ratos
3.
Am J Transplant ; 14(5): 1096-108, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24708486

RESUMO

Transplant ischemia-reperfusion injury (Tx-IRI) and allograft dysfunction remain as two of the major clinical challenges after heart transplantation. We investigated the role of angiopoietin-2 (Ang2) in Tx-IRI and rejection using fully MHC-mismatched rat cardiac allografts. We report that plasma levels of Ang2 were significantly enhanced in the human and rat recipients of cardiac allografts, but not in the rat recipients of syngrafts, during IRI. Ex vivo intracoronary treatment of rat cardiac allografts with anti-Ang2 antibody before 4-h cold preservation prevented microvascular dysfunction, endothelial cell (EC) adhesion molecule expression and leukocyte infiltration, myocardial injury and the development of cardiac fibrosis and allograft vasculopathy. Recipient preoperative and postoperative treatment with anti-Ang2 antibody produced otherwise similar effects without effecting microvascular dysfunction, and in additional experiments prolonged allograft survival. Recipient preoperative treatment alone failed to produce these effects. Moreover, ex vivo intracoronary treatment of allografts with recombinant Ang2 enhanced Tx-IRI and, in an add-back experiment, abolished the beneficial effect of the antibody. We demonstrate that neutralization of Ang2 prevents EC activation, leukocyte infiltration, Tx-IRI and the development of chronic rejection in rat cardiac allografts. Our results suggest that blocking Ang2 pathway is a novel, clinically feasible, T cell-independent strategy to protect cardiac allografts.


Assuntos
Angiopoietina-2/antagonistas & inibidores , Anticorpos Monoclonais/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Transplante de Coração , Complicações Pós-Operatórias/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , Adulto , Idoso , Aloenxertos , Angiopoietina-2/sangue , Angiopoietina-2/imunologia , Animais , Morte Encefálica , Estudos de Casos e Controles , Doença Crônica , Feminino , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/metabolismo , Sobrevivência de Enxerto , Cardiopatias/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/metabolismo , Ratos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Adulto Jovem
4.
Am J Transplant ; 13(8): 2019-34, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23773358

RESUMO

Ischemia-reperfusion injury (IRI) after kidney transplantation may result in delayed graft function. We used rat renal artery clamping and transplantation models to investigate cholesterol-independent effects of clinically relevant single-dose peroral simvastatin treatment 2 h before renal ischemia on microvascular injury. The expression of HMG-CoA reductase was abundant in glomerular and peritubular microvasculature of normal kidneys. In renal artery clamping model with 30-min warm ischemia, simvastatin treatment prevented peritubular microvascular permeability and perfusion disturbances, glomerular barrier disruption, tubular dysfunction and acute kidney injury. In fully MHC-mismatched kidney allografts with 16-h cold and 1-h warm ischemia, donor simvastatin treatment increased the expression of flow-regulated transcription factor KLF2 and vasculoprotective eNOS and HO-1, and preserved glomerular and peritubular capillary barrier integrity during preservation. In vitro EC Weibel-Palade body exocytosis assays showed that simvastatin inhibited ischemia-induced release of vasoactive angiopoietin-2 and endothelin-1. After reperfusion, donor simvastatin treatment prevented microvascular permeability, danger-associated ligand hyaluronan induction, tubulointerstitial injury marker Kim-1 immunoreactivity and serum creatinine and NGAL levels, and activation of innate and adaptive immune responses. In conclusion, donor simvastatin treatment prevented renal microvascular dysfunction and IRI with beneficial effects on adaptive immune and early fibroproliferative responses. Further studies may determine potential benefits in clinical cadaveric kidney transplantation.


Assuntos
Injúria Renal Aguda/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Rim/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Sinvastatina/uso terapêutico , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Animais , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Rim/irrigação sanguínea , Rim/metabolismo , Transplante de Rim , Masculino , Ratos , Ratos Endogâmicos WF , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo
5.
Am J Transplant ; 13(3): 600-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23331861

RESUMO

Ischemia-reperfusion injury (IRI) induces hypoxia-inducible factor-1 (HIF-1) in the myocardium, but the consequences remain elusive. We investigated HIF-1 activation during cold and warm ischemia and IRI in rat hearts and cardiac syngrafts. We also tested the effect of HIF-α stabilizing prolyl hydroxylase inhibitor (FG-4497) on IRI or allograft survival. Ex vivo ischemia of the heart increased HIF-1α expression in a time- and temperature-dependent fashion. Immunohistochemistry localized HIF-1α to all cardiac cell types. After reperfusion, HIF-1α immunoreactivity persisted in smooth muscle cells and cardiomyocytes in the areas with IRI. This was accompanied with a transient induction of protective HIF-1 downstream genes. Donor FG-4497 pretreatment for 4 h enhanced IRI in cardiac allografts as evidenced by an increase in cardiac troponin T release, cardiomyocyte apoptosis, and activation of innate immunity. Recipient FG-4497 pretreatment for 4 h decreased infiltration of ED1(+) macrophages, and mildly improved the long-term allograft survival. In syngrafts donor FG-4497 pretreatment increased activation of innate immunity, but did not induce myocardial damage. We conclude that the HIF-1 pathway is activated in heart transplants. We suggest that pharmacological HIF-α preconditioning of cardiac allografts donors would not lead to clinical benefit, while in recipients it may result in antiinflammatory effects and prolonged allograft survival.


Assuntos
Inibidores Enzimáticos/farmacologia , Transplante de Coração , Coração/fisiopatologia , Fator 1 Induzível por Hipóxia/metabolismo , Precondicionamento Isquêmico , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Inflamação/diagnóstico , Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos WF , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transplante Homólogo
6.
Transplant Proc ; 38(10): 3266-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175244

RESUMO

Obliterative bronchiolitis (OB) is the major limitation for long-term survival of lung allograft recipients. The exact molecular and cellular mechanisms contributing to obliterative lesion formation are unknown. Pathological characteristics of OB are epithelial damage, peribronchial inflammation, and increasing obliteration of bronchioli. Vascular endothelial growth factor (VEGF) is an angiogenic growth factor that exerts proinflammatory effects by increasing endothelial permeability and inducing expression of endothelial adhesion molecules. We investigated the role of VEGF in the development of OB in rat tracheal allografts and the role of VEGF receptors (VEGFR)-1 and -2 in the development of OB in mouse tracheal allografts. In nontreated allografts, with increasing loss of epithelium and airway occlusion, VEGF messenger RNA (mRNA) and protein expression vanished in the epithelium and increased in smooth muscle cells and mononuclear inflammatory cells compared with syngeneic grafts. Intragraft VEGF overexpression by adenoviral transfer of a mouse VEGF164 gene led to a decrease in epithelial necrosis but increased luminal occlusion by >50% compared with AdLacZ-treated rat tracheal allografts. When compared with the control immunoglobulin (Ig)G group, simultaneous treatment with antibodies against VEGFR-1 and -2 significantly lowered the degree of luminal occlusion of mouse tracheal allografts.


Assuntos
Traqueia/transplante , Transplante Homólogo/fisiologia , Transplante Isogênico/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Adenoviridae/genética , Animais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ratos , Ratos Endogâmicos WF , Transplante Heterotópico , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , beta-Galactosidase/genética
7.
Transplant Proc ; 38(10): 3271-3, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175246

RESUMO

BACKGROUND: In chronic rejection, parenchymal fibrosis and cardiac allograft vasculopathy (CAV) characterized by neointimal growth are the leading causes of graft loss for heart transplant recipients. During alloimmune responses a variety of cytokines, adhesion proteins, and growth factors, such as platelet-derived growth factor (PDGF), are up-regulated. The PDGF family (AA, AB, BB, CC, DD), which acts mainly on connective tissue cells, is considered to be a potent mitogenic and chemotactic factor for fibroblasts and vascular smooth muscle cells. In this study, we evaluated the effects of PDGF ligands in chronic rejection. METHODS: Heterotopic heart transplantations were performed between fully major histocompatability complex-mismatched Dark Agouti to Wistar Furth rats receiving cyclosporine immunosuppression. Allograft coronary arteries were perfused with a recombinant adeno-associated virus (AAV) encoding enhanced green fluorescence protein (EGFP) as a control gene or PDGF-A, -B, -C, -D. Allografts were harvested at 100 days for morphometric analysis of CAV and fibrosis. RESULTS: AAV-mediated transgene expression was detected by EGFP immunoreactivity across the graft section (at 100 days) in AAV EGFP-perfused allografts. AAV PDGF-A, -C, and -D perfusion resulted in accelerated CAV and fibrosis. In contrast, AAV PDGF-B perfusion did not induce arteriosclerotic changes or fibrosis in cardiac allografts. CONCLUSIONS: AAV PDGF-A, -C, and -D overexpression accelerated the development of chronic rejection, whereas PDGF-B did not. Our results suggested that more targeted therapy with monoclonal antibodies blocking the active sites of PDGF-A, -C, and -D may produce beneficial effects on heart transplant survival.


Assuntos
Doença das Coronárias/patologia , Transplante de Coração/patologia , Fator de Crescimento Derivado de Plaquetas/genética , Complicações Pós-Operatórias/patologia , Animais , Dependovirus/genética , Fibrose , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WF , Transplante Homólogo , Doenças Vasculares
8.
Transplant Proc ; 38(10): 3372-3, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175275

RESUMO

Hypoxia plays an integral part in cardiac transplantation as prolonged graft preservation is an individual risk factor for the development of cardiac allograft vasculopathy (CAV). In this study we characterized the role of hypoxia-inducible factor-1 (HIF-1) during prolonged graft preservation, ischemia-reperfusion (I/R), acute rejection, and chronic rejection. Heart transplantations were performed from Dark Agouti (DA) to Wister-Furth (allo) or DA to DA (syn) rats, without immunosuppression (acute rejection model, harvested at day 5) or with cyclosporine (chronic rejection model, harvested at day 60). To study the effect of preservation on HIF-1 regulation, normal DA hearts were subjected to different cold ischemia times with or without 45 minutes of additional warm ischemia. The role of I/R was studied by harvesting syngrafts at different time points after reperfusion. Real-time reverse-transcriptase polymerase chain reaction quantified total HIF-1 mRNA, while enzyme-linked immunosorbent assay and immunohistochemistry quantified and localized HIF-1 protein. Our results show that HIF-1 nuclear immunoreactivity is increased during graft preservation and I/R leads to loss of nuclear HIF-1 immunoreactivity. Acute rejection induced HIF-1 in mRNA level. Our findings thus indicated that HIF-1 is activated during transplantation and suggested that manipulation of the HIF-1 pathway might reveal new therapeutic options to manage CAV.


Assuntos
Rejeição de Enxerto/fisiopatologia , Transplante de Coração/fisiologia , Fator 1 Induzível por Hipóxia/genética , Animais , Hipóxia Celular , Regulação da Expressão Gênica , Transplante de Coração/imunologia , Imuno-Histoquímica , Preservação de Órgãos , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WF , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Homólogo , Transplante Isogênico
9.
Am J Respir Crit Care Med ; 164(4): 672-9, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11520736

RESUMO

In this study, the prevention of rat cytomegalovirus (RCMV) infection-enhanced experimental obliterative bronchiolitis in rat tracheal allografts was investigated. RCMV infection markedly enhanced cell proliferation and histological changes of obliterative bronchiolitis, a form of chronic rejection after lung transplantation. These alterations were linked to increased interleukin (IL)-2 and tumor necrosis factor-alpha (TNF-alpha) immunoreactivity, and reduction of IL-10 expression. In recipient rats with acute RCMV infection, prophylaxis with either ganciclovir (DHPG) or hyperimmune serum (HIS) totally prevented RCMV infection-enhanced tracheal occlusion. DHPG treatment initiated during acute RCMV infection also reduced lesion development but markedly less than DHPG prophylaxis. Treatment of acute RCMV infection with HIS alone or in combination with DHPG had no significant effect on tracheal occlusion. Inhibition of the transcription of cytokines by high doses of cyclosporine A significantly reduced RCMV infection-enhanced tracheal obliteration. In rats with chronic RCMV infection, obliterative alterations were prevented by DHPG prophylaxis initiated at the time of transplantation. Prophylaxis either with DHPG or HIS did not affect the amount of infectious RCMV recovered from host salivary glands, nor were there differences seen in RCMV major immediate early DNA expression in tracheal allografts between different antiviral drug regimens. Immunohistochemical analysis of allografts revealed that inhibition of tracheal occlusion by antiviral prophylaxis was associated with a reduction in the number of ED1(+) macrophages and cells staining for Th1 cytokines and TNF-alpha, while immune modulation by cyclosporine A up-regulated IL-10 production. In conclusion, the results of the present study suggest that the CMV infection-enhanced chronic rejection develops independently of viral load but requires both immune activation and simultaneous CMV gene expression beyond immediate early genes.


Assuntos
Antivirais/uso terapêutico , Bronquiolite Obliterante/prevenção & controle , Bronquiolite Obliterante/virologia , Infecções por Citomegalovirus/complicações , Modelos Animais de Doenças , Ganciclovir/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/virologia , Soros Imunes/administração & dosagem , Transplante de Pulmão/efeitos adversos , Pré-Medicação/métodos , Doença Aguda , Animais , Bronquiolite Obliterante/etiologia , Bronquiolite Obliterante/imunologia , Bronquiolite Obliterante/patologia , Doença Crônica , Citomegalovirus/genética , Avaliação Pré-Clínica de Medicamentos , Regulação Viral da Expressão Gênica , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Imuno-Histoquímica , Macrófagos Alveolares/ultraestrutura , Masculino , Ratos , Ratos Endogâmicos , Células Th1/ultraestrutura , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/ultraestrutura , Carga Viral
10.
Transplantation ; 71(12): 1741-7, 2001 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-11455252

RESUMO

BACKGROUND: Studies of cardiac allograft arteriosclerosis, i.e., chronic rejection, have largely focused on mononuclear inflammatory cell infiltrates in the vascular wall and periphery of the occluded vessels. The purpose of this study was to investigate the role of mast cells in the development of acute and chronic rejection in rat cardiac allografts. METHODS: In the acute rejection model, transplant recipients were not treated with immunosuppressants, and the grafts were removed 5 days after transplantation at the time of severe acute rejection. In the chronic rejection model, the recipients were administered triple-drug immunosuppression, and the grafts were removed 90 days after transplantation. RESULTS: During acute rejection, the number of mast cells was not increased, but the localization pattern differed from that of syngeneic grafts. In acute rejection, mast cells were located in the perivascular region of the allografts, but in syngeneic grafts, mast cells had a more interstitial location. In the chronic rejection model, the cardiac allografts with severe intimal thickening showed large numbers of mast cells at perivascular sites of occluded intramyocardial vessels and in the interstitium. Linear regression analysis revealed a significant correlation between the numbers of perivascular and interstitial mast cells and the intensity of intimal thickening. The majority of mast cells showed positive immunoreactivity to basic fibroblast growth factor (bFGF). Macrophage bFGF expression was not so prominent, but macrophages were more frequent in numbers. Tumor necrosis factor-alpha expression was detected mainly in macrophages and in only a few mast cells. When the intensity of arteriosclerosis was decreased by an increase in the intensity of immunosuppression, the numbers of intragraft mast cells and other mononuclear cells, and also the production of their respective cytokines, bFGF and tumor necrosis factor-alpha, gradually diminished. CONCLUSIONS: Taken together, our data show that the intensity of intramyocardial mast cell infiltration was associated with the intensity of chronic inflammation and allograft arteriosclerotic changes, but not with acute rejection, and that mast cells, in addition to macrophages, are a major source of myocardial bFGF. The results also demonstrate that when the T-cell activation pathway is blocked using cyclosporin, the number of mast cells is decreased. Cyclosporin may have affected the cytokine production that interfered with both the mast cell-dependent initiation and the leukocyte- and mast cell-dependent amplification and progression of the immune responses influenced by mast cell-leukocyte cytokine cascades. bFGF produced by mast cells may contribute to enhanced inflammation, neovascularization, and fibrosis during cardiac allograft arteriosclerosis.


Assuntos
Rejeição de Enxerto/patologia , Transplante de Coração , Mastócitos/patologia , Miocárdio/patologia , Doença Aguda , Animais , Doença Crônica , Fator 2 de Crescimento de Fibroblastos/metabolismo , Rejeição de Enxerto/metabolismo , Macrófagos/metabolismo , Miocárdio/metabolismo , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WF , Valores de Referência , Transplante Homólogo , Transplante Isogênico , Fator de Necrose Tumoral alfa/metabolismo
15.
Am J Respir Crit Care Med ; 161(4 Pt 1): 1332-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10764331

RESUMO

The role of complement activation in the development of obliterative bronchiolitis, a manifestation of chronic lung allograft rejection, was investigated in the heterotopic rat tracheal allograft model. An increase in intragraft complement components C3 and C5b-9 (membrane attack complex) as well as IgM and IgG deposits were demonstrated during the progressive loss of respiratory epithelium and airway occlusion in nontreated allografts compared with syngeneic grafts. A 7-d treatment with recombinant human soluble complement receptor type 1 (sCR1; 20 mg/kg/d, intraperitoneal), an inhibitor of both the classic and alternative complement pathways, significantly decreased epithelial necrosis and intragraft neutrophil infiltration, and reduced obliterative changes by 40%. Immunohistochemical analysis of the grafts showed that sCR1 treatment significantly decreased early C5b-9 and IgG deposits, neutrophil chemoattractant IL-8 immunoreactivity, and ICAM-1 expression. Treatment with sCR1 was associated with increased staining for Th2 cytokines, in particular IL-10, with concomitant downregulation of IL-2 and TNF-alpha immunoreactivity. In contrast, sCR1 treatment did not affect the number of graft-infiltrating CD4(+) and CD8(+) T cells, CD45(+) B cells, ED1(+) and ED3(+) macrophages, or immune activation with expression of IL-2Ralpha or MHC class II. In conclusion, this is the first study to demonstrate that blockade of complement activation attenuates the development of OB and suggests that in addition to T cell-driven responses, humoral and antigen-independent immune responses also operate in the disease process. A blockade of complement activation renders the chemokine milieu unattractive to neutrophils and also modulates the alloimmune response toward Th2 cytokines, which may have an antiproliferative role in fibroproliferative disorders.


Assuntos
Bronquiolite Obliterante/prevenção & controle , Ativação do Complemento/fisiologia , Traqueia/transplante , Animais , Bronquiolite Obliterante/imunologia , Quimiocinas/metabolismo , Ativação do Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Citocinas/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Humanos , Imunoglobulina G/metabolismo , Masculino , Omento , Ratos , Ratos Endogâmicos WF , Receptores de Complemento/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Transplante Heterotópico
16.
Cardiovasc Pathol ; 8(6): 339-47, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10615021

RESUMO

Recent evidence supports a role for cell death and inflammation as etiologic factors in neointimal formation and restenosis after angioplasty. This study was undertaken to examine the pattern and intensity of the proliferative response, cell death, and activation of inflammatory, endothelial and smooth muscle cells (SMC) in a model of intimal reinjury. Two ballooning injuries were performed to rat aorta, the second one 14 days after the first injury. Our results demonstrate that ballooning injury to pre-existing neointima differs clearly from an injury to a normal aorta. First, ballooning injury to pre-existing neointima doubled the proliferative response of SMC and intimal thickening, but proliferation of SMC occurred only in the intima, and did not extend into the media. Second, within four hours after the first injury, the number of TUNEL-positive SMC in the media increased from 3% to 23%, but no such increase was found in the pre-existing neointima after the second injury. Third, the prompt proliferative response of intimal SMC after the second injury was linked with a significant increase in endothelial P-selectin and neointimal VCAM-1 immunoreactivity, compared to the first injury at corresponding time points, followed by high numbers of activated ED3+ macrophages and CD4+ T cells in the developing neointima. A balance in injury-induced cell death and proliferation obviously maintains stable cell numbers observed in the media, whereas in the neointima, the resistance of SMC to injury-induced cell death may contribute to a rapid lesion formation in restenosis.


Assuntos
Apoptose , Túnica Íntima/patologia , Animais , Aorta/lesões , Aorta/metabolismo , Aorta/patologia , Bromodesoxiuridina/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Cateterismo , Contagem de Células , Divisão Celular , Modelos Animais de Doenças , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Selectina-P/metabolismo , Ratos , Ratos Wistar , Túnica Íntima/lesões , Túnica Íntima/metabolismo , Túnica Íntima/fisiopatologia , Túnica Média/lesões , Túnica Média/metabolismo , Túnica Média/patologia , Túnica Média/fisiopatologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
17.
Am J Respir Crit Care Med ; 160(4): 1324-32, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508825

RESUMO

The role of platelet-derived growth factor (PDGF) in the development of obliterative bronchiolitis (OB) as a manifestation of chronic rejection was investigated in the heterotopic rat tracheal allograft model. An increase in intragraft PDGF-Ralpha and -Rbeta mRNA expression, and in PDGF-AA and -Ralpha immunoreactivity, was demonstrated during the progressive loss of respiratory epithelium and airway occlusion in nontreated allografts compared with syngeneic grafts. Treatment with CGP 53716, a protein-tyrosine kinase inhibitor selective for PDGF receptor, alone and in combination with suboptimal doses of cyclosporin A, significantly reduced myofibroproliferation and the degree of OB by more than 50%. CGP 53716 did not affect airway wall inflammatory cell proliferation, the number of graft-infiltrating CD4(+) or CD8(+) T cells, ED3(+) macrophages, or the level of immune activation determined as IL-2R and MHC class II expression. This study suggests a regulatory role for PDGF, especially for PDGF-AA and -Ralpha, in the development of obliterative bronchiolitis in this model, and demonstrates that inhibition of PDGF receptor protein-tyrosine kinase activation prevents these obliterative changes. Thus, receptor protein-tyrosine kinase inhibitors may provide a novel therapeutic strategy for the prevention of chronic rejection.


Assuntos
Bronquiolite Obliterante/fisiopatologia , Rejeição de Enxerto/fisiopatologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Animais , Bronquiolite Obliterante/metabolismo , Bronquiolite Obliterante/patologia , Divisão Celular/efeitos dos fármacos , Doença Crônica , Ciclosporina/farmacologia , Inibidores Enzimáticos/farmacologia , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Imuno-Histoquímica , Imunossupressores/farmacologia , Masculino , Reação em Cadeia da Polimerase , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Endogâmicos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Mucosa Respiratória/patologia , Traqueia/metabolismo , Traqueia/patologia , Traqueia/transplante , Transplante Homólogo , Transplante Isogênico
18.
Transpl Infect Dis ; 1(2): 115-26, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11428979

RESUMO

There is a wealth of clinical and experimental evidence indicating the interaction of cytomegalovirus (CMV) infection and rejection in cardiac and other solid organ allografts. A plausible explanation for this association comes from data showing that therapy with biologicals, sepsis, and rejection, all lead to the release of TNF-alpha which, upon binding to its receptor, activates NF-kB. TNF-alpha is also able to stimulate the activity of the CMV-IE enhancer/promoter region. CMV infection of several cell lines leads to NF-kB activation. NF-kB binding sites are present in regulatory regions of various cellular and viral genes, including the IE enhancer region of CMV. In a reciprocal situation, CMV infection, most likely via gamma-interferon, leads to upregulation of MHC antigens in the transplant and, thereby, to increased transplant immunogenicity. Thus, a vicious circle is induced. We have investigated in detail the pathobiology of CMV and allograft vasculopathy (chronic rejection) in experimental animals, using aortic and cardiac allografts as well as a trachea model. The results may be summarized as follows: Infection of the recipient with rat CMV results in an early inflammatory response in the aortic and cardiac allograft vascular adventitia and intima (endothelialitis) and in the airway wall of tracheal allografts. This early inflammatory response leads to enhanced intimal thickness in aortic and cardiac allografts and enhanced luminal occlusion of tracheal allografts. Timewise, this coincides with early activation of intragraft inflammatory leukocytes and increased mRNA of various growth factors and cytokines. When the recipients receive gancyclovir, the enhanced intimal response in aortic and cardiac allografts and luminal occlusion in tracheal allografts is entirely abolished. Gancyclovir treatment dramatically reduces the inflammatory response in the allograft, and thereby growth factor synthesis in response to injury. However, gancyclovir does not prevent the expression of IE antigen of CMV, suggested to inactivate tumor suppressor protein p53 predisposing smooth muscle cells to increased growth. Taken together, the effect of CMV infection on cardiac allograft dysfunction is bidirectional and biphasic. The bidirectional nature emerges from the observations that acute CMV infection may accelerate acute rejection, and, on the other hand, acute alloimmune response-associated cytokine response may activate latent CMV infection. The biphasic effect of CMV on allograft dysfunction refers to its early and late detrimental effects, i.e. during the time of acute and chronic rejection. These two effects of CMV on allograft dysfunction emphasize the need for precise diagnosis of CMV infection in transplant recipients and pre-emptive or prophylactic anti-viral therapy. The benefits of this strategy may not be evident during the early post-transplant period, but 5-10 years after transplantation they manifest as better graft survival.


Assuntos
Vasos Coronários/patologia , Infecções por Citomegalovirus/fisiopatologia , Citomegalovirus/genética , Transplante de Coração/patologia , Complicações Pós-Operatórias , Animais , Arteriosclerose/patologia , Arteriosclerose/virologia , Doença das Coronárias/patologia , Doença das Coronárias/virologia , Vasos Coronários/virologia , Infecções por Citomegalovirus/imunologia , Humanos , Ratos , Transplante Homólogo
19.
Circulation ; 96(4): 1240-9, 1997 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-9286955

RESUMO

BACKGROUND: The molecular mechanisms of cardiac allograft vasculopathy (CAV) remain largely unknown. Using rat cardiac allografts, we examined by immunohistochemistry the expression and localization of platelet-derived growth factor ligand (PDGF-AA and -BB) and receptor (R alpha and R beta) proteins during acute and chronic rejection. METHODS AND RESULTS: In acute rejection, a prominent induction of both PDGF ligand and receptor proteins occurred in the interstitial mononuclear inflammatory cells (P<.05), most of which were ED1-immunoreactive. PDGF-R beta was also induced in the capillary endothelium (P<.01). In cardiac allografts with severe intimal thickening, PDGF-AA expression was localized to the media and intima, whereas PDGF-BB expression was less prominent and was detected mainly in interstitial ED1-immunoreactive inflammatory cells. Double staining revealed that intimal cells expressing PDGF-AA were alpha-smooth muscle actin-positive but also alpha-smooth muscle actin-negative myofibroblast-like cells and to a lesser extent, ED1-immunoreactive cells. Both PDGF-R alpha and -R beta expression occurred in intimal, arterial endothelial, and interstitial mononuclear inflammatory cells. High-dose cyclosporin A (CsA) treatment significantly reduced both PDGF-AA and PDGF-R alpha expression in intimal cells. Furthermore, linear regression analysis revealed that PDGF-AA, PDGF-R alpha, and PDGF-R beta expression in intimal cells and PDGF-BB expression in interstitial mononuclear inflammatory cells correlated with intimal thickening. CONCLUSIONS: Alloimmune injury induces the expression of PDGF ligands, especially of PDGF-AA, in the graft vasculature and sufficient immunosuppression with CsA suppresses the expression of PDGF and inhibits the development of CAV. PDGF may have a substantial role in the regulation of smooth muscle cell migration and proliferation in an autocrine or paracrine manner during the development of CAV.


Assuntos
Rejeição de Enxerto/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Doença Aguda , Animais , Doença Crônica , Rejeição de Enxerto/patologia , Terapia de Imunossupressão , Ligantes , Fator de Crescimento Derivado de Plaquetas/análise , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WF , Receptores do Fator de Crescimento Derivado de Plaquetas/análise
20.
Am J Respir Crit Care Med ; 155(6): 2078-88, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9196118

RESUMO

Inbred DA (AG-B4, RT1a) and WF (AG-B2, RT1v) rat immunosuppressed with cyclosporine A (CsA) 2 mg/kg/day subcutaneously were used as donors and recipients of heterotopic rat tracheal allografts. Acute infection was established by inoculating the recipients with 10(5) plaque-forming units of rat cytomegalovirus (RCMV) intraperitoneal on the day of transplantation. Chronic RCMV infection was similarly established 8 wk before transplantation in donors alone, recipients alone, and in both donor and recipients. The control rats were left noninfected. For in vivo cell proliferation, the rats received bromedeoxyuridine intravenously 3 h before being killed. RCMV infection significantly enhanced the generation of experimental obliterative bronchiolitis (OB). First, acute RCMV infection was linked to markedly enhanced MHC class II expression on the respiratory epithelium and prominent airway wall inflammation of W3/25+ T cells (CD4+) and ED1+/ED2+ macrophages. Second, acute infection induced a fivefold increase in luminal occlusion of the trachea, due to proliferating inflammatory and alpha-smooth muscle actin+ myofibroblast-like cells. Acute RCMV infection was particularly associated with markedly increased expression of platelet-derived growth factor (PDGF) AA-isoform in various structures of the graft 10 d after transplantation. At 30 d, RCMV significantly increased PDGF alpha- and beta-receptor expression in alpha-smooth muscle actin+ cells and TNF-alpha expression in capillary endothelial cells of the fibroproliferative lesion. In chronic recipient RCMV infection, the histopathological changes were quite similar to acute infection. Chronic donor infection did not amplify the development of experimental OB. As analyzed by immunohistochemistry from the grafts and by plaque assays from salivary gland biopsies of the recipients, trace RCMV antigen expression, while no infectious RCMV could be recovered in the chronic donor infection group. In other infected groups, RCMV antigen expression was mild to moderate and salivary glands contained plenty of infectious RCMV. To conclude, our results indicate that RCMV infection enhances epithelial MHC class II expression and myofibroproliferation in heterotopic rat tracheal allografts and suggest that these changes may be induced by increased PDGF-AA and PDGF alpha-receptor expression, respectively.


Assuntos
Bronquiolite Obliterante/complicações , Infecções por Citomegalovirus/complicações , Traqueia/transplante , Traqueia/virologia , Doenças da Traqueia/complicações , Doença Aguda , Animais , Antígenos Virais/metabolismo , Doença Crônica , Citocinas/metabolismo , Infecções por Citomegalovirus/imunologia , Substâncias de Crescimento/metabolismo , Complicações Pós-Operatórias , Ratos , Doadores de Tecidos , Traqueia/metabolismo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...