Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Pathol ; 185(4): 927-42, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25794704

RESUMO

Macrophages are increasingly recognized as a potential therapeutic target in myocardial fibrosis via interactions with fibroblasts. We have characterized macrophage depletion and inhibition of nonclassical macrophage migration, in addition to direct interactions between nonclassical macrophages and fibroblasts in angiotensin II (AngII)-mediated, hypertensive myocardial fibrosis. Macrophage depletion was achieved by daily i.v. clodronate liposomes (-1 day to +3 days) during AngII infusion. Cx3cr1(-/-) mice were used to inhibit nonclassical macrophage migration. Macrophage phenotype (F4/80, CD11b, Ly6C) was characterized by immunofluorescence and flow cytometry. Collagen was assessed by Sirius Red/Fast Green. Quantitative real-time RT-PCR was performed for transcript levels. AngII/wild-type (WT) mice displayed significant infiltrate and fibrosis compared with saline/WT, which was virtually ablated by clodronate liposomes independent of hypertension. In vitro data supported M2 macrophages promoting fibroblast differentiation and collagen production. AngII/Cx3cr1(-/-) mice, however, significantly increased macrophage infiltrate and fibrosis relative to AngII/WT. AngII/Cx3cr1(-/-) mice also showed an M1 phenotypic shift relative to WT mice in, which the predominant phenotype was Ly6C(low), CD206(+) (M2). Myocardial IL-1ß was significantly up-regulated, whereas transforming growth factor ß down-regulated with this M1 shift. We demonstrated that infiltrating macrophages are critical to AngII-mediated myocardial fibrosis by preventing the development of fibrosis after liposomal depletion of circulating monocytes. Our findings also suggest that some macrophages, namely M2, may confer a protective myocardial environment that may prevent excessive tissue injury.


Assuntos
Macrófagos/metabolismo , Miocárdio/patologia , Actinas/metabolismo , Administração Intravenosa , Angiotensina II/administração & dosagem , Angiotensina II/farmacologia , Animais , Antígenos Ly/metabolismo , Receptor 1 de Quimiocina CX3C , Ácido Clodrônico/administração & dosagem , Ácido Clodrônico/farmacologia , Colágeno/biossíntese , Eletrocardiografia , Fibrose , Mediadores da Inflamação/metabolismo , Lipossomos/administração & dosagem , Lipossomos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Miocárdio/metabolismo , Células NIH 3T3 , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/metabolismo
2.
J Heart Lung Transplant ; 34(5): 734-45, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25655346

RESUMO

BACKGROUND: Cardiac allograft vasculopathy (CAV) is a multifactorial pathology limiting the survival of cardiac transplants. The etiology of CAV is unclear, but antibody-mediated and cellular-mediated responses have been implicated. We, and others, have observed ectopic lymphoid structures (ELS) surrounding epicardial coronary arteries with CAV. The potential contribution of these ELS to CAV has not been elucidated. METHODS: Epicardial coronary arteries were collected from 59 transplant patients at 2 centers and studied for ELS presence and composition using immunohistochemistry. The intima and ELS were isolated, and the expression of the genes involved in tertiary lymphoid organ (TLO) formation was measured by quantitative polymerase chain reaction. RESULTS: ELS presence was related to survival after transplantation (p = 0.013) and histologic composition of CAV (p < 0.001). ELS contain B and T lymphocytes, macrophages, and antibody-producing (immunoglobulin [Ig] M and/or IgG) plasma cells. A sub-population of B lymphocytes appeared to be cluster of differentiation (CD)20(+)CD27(+) memory B lymphocytes. The messenger RNA expression of TLO markers (lymphotoxin-ß, and chemokine [C-C motif] ligand 19 and 21) was significantly higher in ELS than in the neointimal lesions. The ELS observed in this study exhibited some TLO markers but did not exhibit the distinct areas rich in B and T lymphocytes that are normally found in classic TLOs. CONCLUSIONS: The cellular composition of the ELS differs from the cellular infiltrate in CAV intimal lesions. The presence of memory B lymphocytes and plasma producing IgM and IgG cells suggests that ELS are related to local antibody production, potentially contributing to antibody-mediated CAV. ELS associated with coronary vessels containing CAV show features of underdeveloped TLOs; classic TLOs may not develop due to patient immunosuppression.


Assuntos
Vasos Coronários/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração , Imunidade Celular , Tecido Linfoide/imunologia , Linfócitos T/imunologia , Adulto , Aloenxertos , Vasos Coronários/patologia , Endotélio Vascular , Feminino , Rejeição de Enxerto/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Adulto Jovem
3.
Am J Pathol ; 185(3): 631-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25701883

RESUMO

Heart failure, the leading cause of hospitalization of elderly patients, is correlated with myocardial fibrosis (ie, deposition of excess extracellular matrix proteins such as collagen). A key regulator of collagen homeostasis is lysyl oxidase (LOX), an enzyme responsible for cross-linking collagen fibers. Our objective was to ameliorate age-related myocardial fibrosis by disrupting collagen cross-linking through inhibition of LOX. The nonreversible LOX inhibitor ß-aminopropionitrile (BAPN) was administered by osmotic minipump to 38-week-old C57BL/6J male mice for 2 weeks. Sirius Red staining of myocardial cross sections revealed a reduction in fibrosis, compared with age-matched controls (5.84 ± 0.30% versus 10.17 ± 1.34%) (P < 0.05), to a level similar to that of young mice at 8 weeks (4.9 ± 1.2%). BAPN significantly reduced COL1A1 mRNA, compared with age-matched mice (3.5 ± 0.3-fold versus 15.2 ± 4.9-fold) (P < 0.05), suggesting that LOX is involved in regulation of collagen synthesis. In accord, fibrotic factor mRNA expression was reduced after BAPN. There was also a novel increase in Ly6C expression by resident macrophages. By interrupting collagen cross-linking by LOX, the BAPN treatment reduced myocardial fibrosis. A novel observation is that BAPN treatment modulated the transforming growth factor-ß pathway, collagen synthesis, and the resident macrophage population. This is especially valuable in terms of potential therapeutic targeting of collagen regulation and thereby age-related myocardial fibrosis.


Assuntos
Aminopropionitrilo/uso terapêutico , Colágeno/metabolismo , Cardiopatias/tratamento farmacológico , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Fatores Etários , Aminopropionitrilo/farmacologia , Animais , Fibrose/metabolismo , Fibrose/patologia , Cardiopatias/metabolismo , Cardiopatias/patologia , Masculino , Camundongos , Miocárdio/patologia
4.
J Heart Lung Transplant ; 32(4): 454-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23498165

RESUMO

BACKGROUND: Epicardial cardiac allograft vasculopathy (CAV) is commonly described as a homogeneous smooth muscle cell (SMC)-rich inward intimal lesion with the SMC oriented circumferentially around the vessel. Recent findings have called this description into question. In this study we aimed to clarify the clinical presentation of epicardial CAV. METHODS: Autopsied samples of the 3 major coronaries were analyzed from patients fitting cardiac donor criteria (n = 10) and patients who had undergone cardiac transplantation (n = 34). Histology and immunohistochemistry were performed to identify cellular components of CAV, and image analysis was used to measure the various vascular compartments. RESULTS: Of the 34 cases examined, 28 of the epicardial intimal lesions contained 2 clearly definable layers overlying the media. The layer most adjacent to the media was SMC-rich, with the SMC oriented longitudinally along the vessel length and containing few macrophages, both characteristics of donor-derived benign intimal thickening (BIT). Transplants harvested at 1, 4 or 10 days post-transplant confirmed retention of BIT after transplantation. Image analysis of later transplants supported a hypothesis of carry-over BIT in CAV. The more lumenal CAV layer more closely resembled naturally occurring atherosclerosis. CONCLUSIONS: We propose that retention of the SMC-rich BIT layer after transplantation accounts, to a large extent, for the donor-derived, SMC-rich nature of human CAV, and that perturbation of the BIT provides the inflammatory foundation for the development of an accelerated atherosclerosis in the epicardial coronaries of transplant patients. This expanding accelerated atherosclerosis along with the underlying BIT demonstrates the characteristics ascribed to CAV.


Assuntos
Transplante de Coração/efeitos adversos , Túnica Íntima/patologia , Doenças Vasculares/etiologia , Doenças Vasculares/patologia , Adulto , Feminino , Humanos , Pessoa de Meia-Idade , Doadores de Tecidos , Transplante Homólogo/efeitos adversos
5.
Am J Pathol ; 182(3): 714-26, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23287510

RESUMO

Exposure of rodents to angiotensin II (AngII) is a common model of fibrosis. We have previously shown that cellular infiltration of bone marrow-derived progenitor cells (fibrocytes) occurs before deposition of extracellular matrix and is associated with the production of connective tissue growth factor (CTGF). In the present study, we characterized the role of CTGF in promoting fibrocyte accumulation and regulation after AngII exposure. In animals exposed to AngII using osmotic minipumps (2.0 µg/kg per min), myocardial CTGF mRNA peaked at 6 hours (21-fold; P < 0.01), whereas transforming growth factor-ß (TGF-ß) peaked at 3 days (fivefold; P < 0.05) compared with saline control. Early CTGF expression occurred before fibrocyte migration (1 day) into the myocardium or ECM deposition (3 days). CTGF protein expression was evident by day 3 of AngII exposure and seemed to be localized to resident cells. Isolated cardiomyocytes and microvascular endothelial cells responded to AngII with increased CTGF production (2.1-fold and 2.8-fold, respectively; P < 0.05), which was abolished with the addition of anti-TGF-ß neutralizing antibody. The effect of CTGF on isolated fibrocytes suggested a role in fibrocyte proliferation (twofold; P < 0.05) and collagen production (2.3-fold; P < 0.05). In summary, we provide strong evidence that AngII exposure first resulted in Smad2-dependent production of CTGF by resident cells (6 hours), well before the accumulation of fibrocytes or TGF-ß mRNA up-regulation. In addition, CTGF contributes to fibrocyte proliferation in the myocardium and enhances fibrocyte differentiation into a myofibroblast phenotype responsible for ECM deposition.


Assuntos
Angiotensina II/farmacologia , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Fator de Crescimento do Tecido Conjuntivo/farmacologia , Citocinas/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Crescimento Transformador beta/metabolismo
6.
PLoS One ; 7(9): e45663, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029168

RESUMO

AIMS: Myocardial fibrosis contributes to the development of heart failure. Activated Protein C (aPC) is a circulating anticoagulant with anti-inflammatory and cytoprotective properties. Using a model of myocardial fibrosis second to Angiotensin II (AngII) infusion, we investigated the novel therapeutic function aPC in the development of fibrosis. METHODS AND RESULTS: C57Bl/6 and Tie2-EPCR mice were infused with AngII (2.0 µg/kg/min), AngII and aPC (0.4 µg/kg/min) or saline for 3d. Hearts were harvested and processed for analysis or used for cellular isolation. Basic histology and collagen deposition were assessed using histologic stains. Transcript levels of molecular mediators were analyzed by quantitative RT-PCR. Mice infused with AngII exhibited multifocal areas of myocardial cellular infiltration associated with significant collagen deposition compared to saline control animals (p<0.01). AngII-aPC infusion inhibited this cellular infiltration and the corresponding collagen deposition. AngII-aPC infusion also inhibited significant expression of the pro-fibrotic cytokines TGF-ß1, CTGF and PDGF found in AngII only infused animals (p<0.05). aPC signals through its receptor, EPCR. Using Tie2-EPCR animals, where endothelial cells over-express EPCR and exhibit enhanced aPC-EPCR signaling, no significant reduction in cellular infiltration or fibrosis was evident with AngII infusion suggesting aPC-mediate protection is endothelial cell independent. Isolated infiltrating cells expressed significant EPCR transcripts suggesting a direct effect on infiltrating cells. CONCLUSIONS: This data indicates that aPC treatment abrogates the fibrogenic response to AngII. aPC does not appear to confer protection by stimulating the endothelium but by acting directly on the infiltrating cells, potentially inhibiting migration or activation.


Assuntos
Angiotensina II/administração & dosagem , Modelos Animais de Doenças , Fibrose/prevenção & controle , Cardiopatias/prevenção & controle , Proteína C/farmacologia , Animais , Sequência de Bases , Citocinas/metabolismo , Primers do DNA , Fibrose/metabolismo , Imunofluorescência , Cardiopatias/metabolismo , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Heart Lung Transplant ; 31(8): 874-80, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22554675

RESUMO

BACKGROUND: Late cardiac graft rejection, primarily mediated by allograft vasculopathy (AV), remains a major limitation to cardiac transplantation, even in the face of significant calcineurin inhibitor (CNI) immunosuppression. The role played by alloantibody in AV is unclear. Evidence that CNI immunosuppression suppresses CD4(+) T-cell function would suggest that antibody production and effector function would be severely limited in CNI-treated patients. In this study we examine the capacity of CNI-treated animals to develop effective alloantibody that can mediate AV. METHODS: Wild-type (WT) B6 mice were alloimmunized using donor splenocytes or a fully major histocompatibility complex-mismatched allogeneic abdominal aortic graft in the presence of CNI immunosuppression (30 or 50 mg/kg/day cyclosporine A). Anti-serum was harvested and tested using complement-dependent in vitro cytotoxicity assays. Anti-serum was passively transferred to immunodeficient RAG1(-/-) recipients of allogeneic grafts. C4d deposition was quantified in the allografts from WT recipients. RESULTS: CNI immunosuppression did not prevent the development of alloantibody in response to either immunization method (p < 0.05). Passive transfer of anti-serum generated AV lesions in immunodeficient graft recipients and mediated complement-dependent destruction of donor cells (p < 0.05). C4d deposition was localized to the media of grafts of CNI treated animals. CONCLUSIONS: CNI therapy does not prevent the production of alloantibody with the capacity to mediate AV. C4d deposition in the media suggests a role for medial smooth muscle cell loss in antibody-mediated AV lesion development in our model.


Assuntos
Especificidade de Anticorpos/imunologia , Ciclosporina/uso terapêutico , Transplante de Coração , Imunossupressores/uso terapêutico , Isoanticorpos/metabolismo , Doadores de Tecidos , Doenças Vasculares/imunologia , Animais , Inibidores de Calcineurina , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Modelos Animais , Linfócitos T/metabolismo , Transplante Homólogo , Doenças Vasculares/complicações
8.
Int J Exp Pathol ; 93(2): 115-24, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22225615

RESUMO

Using an established model of myocardial hypertrophy and fibrosis after angiotensin II (AngII) infusion, our aim was to characterize the early cellular element involved in the development of myocardial fibrosis in detail. Male Lewis rats were infused with saline or AngII (0.7 mg/kg per day) for up to seven days. Collagen deposition and cellular infiltration were identified by histology stains. Infiltrating cells were grown in vitro and examined by flow cytometry and immunostaining. Chemokine expression was measured using qRT-PCR. AngII infusion resulted in multifocal myocardial cellular infiltration (peak at three days) that preceded collagen deposition. Monocyte chemotactic protein (MCP)-1 transcripts peaked after one day of AngII exposure. Using a triple-labelling technique, the infiltrating cells were found to express markers of leucocyte (ED1(+)), mesenchymal [α-smooth muscle actin (SMA)(+)] and haematopeotic progenitor cells (CD133(+)) suggesting a fibroblast progenitor phenotype. In vitro, ED1(+)/SMA(+)/CD133(+) cells were isolated and grown from AngII-exposed animals. Comparatively few cells were cultured from untreated control hearts, and they were found to be ED1(-)/SMA(+)/CD133(-). We provide evidence that myocardial ECM deposition is preceded by infiltration into the myocardium by cells that express a combination of haematopoietic (ED1, CD133) and mesenchymal (SMA) cell markers, which is a characteristic of the phenotype of fibroblast precursor cells, termed fibrocytes. This suggests that fibrocytes rather than (as is often presumed) leucocytes may have effector functions in the initiation of myocardial fibrosis.


Assuntos
Angiotensina II/toxicidade , Fibroblastos/patologia , Células-Tronco Mesenquimais/patologia , Miocárdio/patologia , Antígeno AC133 , Actinas/metabolismo , Animais , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Ectodisplasinas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Glicoproteínas/metabolismo , Coração/efeitos dos fármacos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Peptídeos/metabolismo , Ratos , Ratos Endogâmicos Lew
9.
J Cardiothorac Surg ; 6: 2, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-21211039

RESUMO

BACKGROUND: Ischemia/reperfusion induced innate immune injury is inescapable in solid organ transplantation. Prolonged cold ischemia exacerbates the primary manifestation of late graft rejection, allograft vasculopathy (AV). The relationship between prolonged cold ischemia and late graft events is unclear and the subject of this study. METHODS: Aortic interposition transplants were performed between fully disparate mice treated with CyclosporineA. Allografts were exposed to 20 min or 60 min of cold ischemia and harvested between 1 d-6 wk. Lesion size, smooth muscle cells (SMC), neutrophils (NØ), and CD8+ T cells were quantified. RESULTS: Early SMC loss was identical in both groups. When compared to 20 min cold ischemia, grafts exposed to 60 min exhibited greater early NØ influx, greater SMC proliferation but fewer medial SMC at 1 wk and 2 wk. Subsequently, earlier and greater CD8+ T cell infiltration were seen in the 60 min group with larger lesions at every time point. CONCLUSIONS: These data suggest that the larger neointimal lesions in grafts exposed to 60 min cold ischemia result from enhanced early innate immune events resulting in impaired SMC recovery and subsequent increased adaptive immune response.


Assuntos
Aorta Abdominal/transplante , Doenças da Aorta/imunologia , Isquemia Fria/efeitos adversos , Imunidade Inata , Músculo Liso Vascular/patologia , Traumatismo por Reperfusão/imunologia , Doenças Vasculares/imunologia , Animais , Doenças da Aorta/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/imunologia , Traumatismo por Reperfusão/patologia , Fatores de Tempo , Doenças Vasculares/patologia
10.
Lab Invest ; 91(4): 565-78, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21116240

RESUMO

Myocardial fibrosis is characterized by significant extracellular matrix (ECM) deposition. The specific cellular mediators that contribute to the development of fibrosis are not well understood. Using a model of fibrosis with Angiotensin II (AngII) infusion, our aim was to characterize the cellular elements involved in the development of myocardial fibrosis. Male C57Bl/6 and Tie2-GFP mice were given AngII (2.0 mg/kg/min) or saline (control) via mini osmotic pumps for up to 7 days. Hearts were harvested, weighed and processed for analysis. Cellular infiltration and collagen deposition were quantified. Immunostaining was performed for specific markers of leukocytes (CD45, CD11b), myofibroblasts (SMA), endothelial cells (vWF) and hematopoietic progenitor cells (CD133). Bone marrow (BM) origin of infiltrating cells was assessed using GFP(+) chimeric animals. Relative qRT-PCR was performed for pro-fibrotic cytokines (transforming growth factor (TGF)-ß1, CTGF) as well as the chemokine stromal-derived factor (SDF)-1α. Myocardial-infiltrating cells were grown in vitro. AngII exposure resulted in multifocal myocardial cellular infiltration, which preceded extensive ECM deposition. A limited number of myocardial-infiltrating cells were positive for leukocyte markers but were significantly positive for myofibroblast (SMA) and endothelial cell (vWF) markers. However, using Tie2-GFP mice, where endothelial cells are GFP(+), myocardial-infiltrating cells were not GFP(+). Transcript levels for SDF-1α were significantly elevated at 1 day of AngII exposure suggesting that hematopoietic progenitor cells may be recruited. This was confirmed by positive CD133 staining of infiltrating cells and evident GFP(+) cellular infiltration when exposing GFP(+) BM chimeras to AngII. Furthermore, a significant number of CD133(+)/SMA(+) cells were grown in vitro from the myocardium of AngII-exposed animals (P<0.01). Myocardial ECM deposition is preceded by the infiltration of the myocardium with hematopoietic cells that express mesenchymal markers. These data suggest that mesenchymal progenitor cells are recruited, and may have a primary role, in the initiation of myocardial fibrosis.


Assuntos
Angiotensina II/administração & dosagem , Células-Tronco Mesenquimais/patologia , Miocárdio/patologia , Antígeno AC133 , Actinas/metabolismo , Animais , Antígenos CD/metabolismo , Células da Medula Óssea/metabolismo , Divisão Celular , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimera , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibrose , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Infusões Subcutâneas , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso/metabolismo , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Fator de von Willebrand/metabolismo
11.
Can J Microbiol ; 56(6): 487-94, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20657619

RESUMO

Multidrug-resistant Mycobacterium tuberculosis strains have rapidly become a global health concern. North American First Nations communities have used traditional medicines for generations to treat many pulmonary infections. In this study, we evaluated the antimycobacterial activity of 5 medicinal plants traditionally used as general therapeutics for pulmonary illnesses and specifically as treatments for tuberculosis. Aqueous extracts of Aralia nudicaulis, Symplocarpus foetidus, Heracleum maximum, Juniperus communis, and Acorus calamus were screened for antimycobacterial activity against Bacillus Calmette-Guérin, Mycobacterium avium, and M. tuberculosis H37Ra using the colorimetric microplate resazurin assay. Extracts of Acorus calamus and H. maximum root demonstrated significant antimycobacterial activity comparable to that of the rifampin control (2 microg/mL). Evaluation of the cytotoxicity of these 2 extracts using the MTT assay also showed that the extracts were less toxic to 3 human cell lines than was the DMSO positive control. This study demonstrates that aqueous extracts of the roots of H. maximum and Acorus calamus possess strong in vitro antimycobacterial activity, validates traditional knowledge, and provides potential for the development of urgently needed novel antituberculous therapeutics.


Assuntos
Antituberculosos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Mycobacterium avium/efeitos dos fármacos , Mycobacterium bovis/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Extratos Vegetais/farmacologia , Plantas Medicinais/química , Antituberculosos/isolamento & purificação , Humanos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Mycobacterium avium/metabolismo , Mycobacterium bovis/metabolismo , Mycobacterium tuberculosis/metabolismo , Oxazinas/metabolismo , Extratos Vegetais/isolamento & purificação , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo , Xantenos/metabolismo
12.
J Cardiothorac Surg ; 5: 52, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20569484

RESUMO

BACKGROUND: Cardiac allograft vasculopathy (AV) is a pathological process of vascular remodeling leading to late graft loss following cardiac transplantation. While there is consensus that AV is alloimmune mediated, and evidence that the most important alloimmune target is medial smooth muscle cells (SMC), the role of the innate immune response in the initiation of this disease is still being elucidated. As ischemia reperfusion (IR) injury plays a pivotal role in the initiation of AV, we hypothesize that IR enhances the early innate response to cardiac allografts. METHODS: Aortic transplants were performed between fully disparate mouse strains (C3H/HeJ and C57BL/6), in the presence of therapeutic levels of Cyclosporine A, as a model for cardiac AV. Neutrophils were depleted from some recipients using anti-PMN serum. Grafts were harvested at 1,2,3,5d and 1,2wk post-transplant. Ultrastructural integrity was examined by transmission electron microscopy. SMC and neutrophils were quantified from histological sections in a blinded manner. RESULTS: Grafts exposed to cold ischemia, but not transplanted, showed no medial SMC loss and normal ultrastructural integrity. In comparison, allografts harvested 1d post-transplant exhibited > 90% loss of SMC (p < 0.0001). SMC partially recovered by 5d but a second loss of SMC was observed at 1wk. SMC loss at 1d and 1wk post-transplant correlated with neutrophil influx. SMC loss was significantly reduced in neutrophil depleted recipients (p < 0.01). CONCLUSIONS: These novel data show that there is extensive damage to medial SMC at 1d post-transplant. By depleting neutrophils from recipients it was demonstrated that a portion of the SMC loss was mediated by neutrophils. These results provide evidence that IR activation of early innate events contributes to the etiology of AV.


Assuntos
Transplante de Coração/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Neutrófilos/imunologia , Doenças Vasculares/imunologia , Animais , Modelos Animais de Doenças , Transplante de Coração/efeitos adversos , Transplante de Coração/patologia , Imunidade Inata/imunologia , Masculino , Camundongos , Miócitos de Músculo Liso/patologia , Transplante Homólogo
13.
J Thorac Cardiovasc Surg ; 140(4): 801-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20176369

RESUMO

OBJECTIVE: The study objectives were to assess the efficacy of N,O carboxymethyl chitosan film in reducing postsurgical adhesion in a rabbit cardiac injury model and to confirm the efficacy of N,O carboxymethyl chitosan gel and film in reducing postsurgical adhesion formation in a pig cardiac injury model. METHODS: (1) Rabbit cardiac injury model: Cardiac injury was generated by abrading the anterior surface of the heart and desiccation with oxygen. N,O carboxymethyl chitosan solution and film were administered to the injured surface. (2) Pig cardiac injury model: Cardiac injury was generated as described above. N,O carboxymethyl chitosan solution and gel (or film) were administered to the injured surface. The severity and area of adhesion between the heart and the sternum were evaluated at 14 days postcardiac surgery. RESULTS: (1) Rabbits treated with N,O carboxymethyl chitosan film plus solution showed significantly reduced severity and area of adhesion formation. (2) Both N,O carboxymethyl chitosan gel plus solution and N,O carboxymethyl chitosan film plus solution significantly reduced adhesion formation in the pig model. CONCLUSIONS: Application of N,O carboxymethyl chitosan products significantly reduces severity of postsurgical adhesion formation after cardiac surgery in the rabbit and pig models. N,O carboxymethyl chitosan products may act as a biophysical barrier.


Assuntos
Materiais Biocompatíveis , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Quitosana/farmacologia , Cardiopatias/prevenção & controle , Administração Tópica , Animais , Química Farmacêutica , Quitosana/administração & dosagem , Quitosana/química , Modelos Animais de Doenças , Feminino , Géis , Cardiopatias/etiologia , Cardiopatias/patologia , Masculino , Coelhos , Índice de Gravidade de Doença , Suínos , Fatores de Tempo , Aderências Teciduais
14.
Transpl Immunol ; 22(3-4): 157-64, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19895889

RESUMO

Using a clinically relevant, fully disparate, allogeneic aortic transplant mouse model of allograft vasculopathy, we have demonstrated that neointimal proliferation is dependent on CD8(+) T cell effector pathways in the presence of therapeutic doses of calcineurin inhibitor (CNI) immunosuppression. CD4(+) T cell pathways are ablated by CNI immunosuppression. In the current study, we examined the relationship between CD8(+) T cell activities, medial SMC loss and neointimal hyperplasia. We demonstrate that at 5-6wk post transplantation in a wild type/wild type transplant CD8(+) T cell infiltration, CD8(+) CTL effector cell mediator expression and medial SMC loss all occur within aortic interposition grafts in the face of CNI immunosuppression. Both IFN-gamma and CTL mediated effector function is required for SMC loss and lesion formation under these conditions. Using strain combinations and reconstitution models, we provide data that blockade of the perforin/granzyme pathway does not prevent lesion formation but that blockade of the Fas/FasL pathway of cytotoxicity dramatically reduces SMC loss and prevents neointimal lesion formation. Both of these blockade strategies are in the face of an active IFN-gamma pathway. These data suggest a cooperative role between Fas/FasL and IFN-gamma mediated effector functions in medial SMC loss and neointimal lesion formation.


Assuntos
Aorta/imunologia , Proteína Ligante Fas/metabolismo , Oclusão de Enxerto Vascular/imunologia , Interferon gama/metabolismo , Receptor fas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Aorta/transplante , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Calcineurina/administração & dosagem , Movimento Celular , Citotoxicidade Imunológica/efeitos dos fármacos , Oclusão de Enxerto Vascular/tratamento farmacológico , Oclusão de Enxerto Vascular/patologia , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Terapia de Imunossupressão , Peptídeos e Proteínas de Sinalização Intracelular , Cooperação Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Fosfoproteínas/administração & dosagem , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/imunologia , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Túnica Média/imunologia , Túnica Média/metabolismo , Túnica Média/patologia
15.
Transpl Immunol ; 22(1-2): 93-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19632325

RESUMO

Allograft vasculopathy (AV) has emerged as a major obstacle for long-term graft survival after cardiac transplantation. The shortage of donor hearts has meant fewer restrictions have been placed on acceptable hearts over the past few years resulting in an increase in the number of older hearts in the donor pool. This increase has subsequently led to the increase of donor hearts containing pre-existing disease. The importance of this pre-existing donor vascular disease in AV outcomes remains controversial. In this study we address this by taking advantage of the fact that B6 Apolipoprotein-E knockout mice develop atherosclerotic lesions in their aortic tracts that closely model human naturally occurring vascular disease. By using these mice as donors, we transplant known levels of pre-existing disease into fully disparate (C3H) recipients. Cyclosporin A is used to prevent acute rejection and allow for allograft vasculopathy. We found that pre-existing lesions are retained in this model after transplantation and that they contribute to increase in lesion size and to increased lumenal narrowing. The de novo AV lesions overlay the pre-existing lesions and this leads to areas of eccentric lesion formation in the vessels with likely accompanying exacerbation of flow perturbation.


Assuntos
Aorta/patologia , Aorta/transplante , Doadores de Tecidos , Doenças Vasculares/etiologia , Doenças Vasculares/patologia , Animais , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Linfócitos T CD4-Positivos/patologia , Ciclosporina/uso terapêutico , Modelos Animais de Doenças , Células Espumosas/patologia , Rejeição de Enxerto/prevenção & controle , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transplante Homólogo/patologia , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Túnica Média/metabolismo , Túnica Média/patologia
16.
Immunopharmacol Immunotoxicol ; 30(3): 553-74, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18618312

RESUMO

Public interest in Echinacea is growing rapidly. Unfortunately, there is little scientific evidence to support claims of efficacy of this widely used botanical, and little information about potential mechanism of action. This study examines the ability of Echinacea to upregulate macrophage function and begins to elucidate the mechanism of Echinacea-induced macrophage activation. Murine peritoneal macrophages were cultured with E. purpurea extracts enriched for plant polysaccharide (EP). ELISA was used to measure cytokine production. MAPKs were blocked using specific inhibitors, and Western blotting used to identify phosphorylated proteins involved in signal transduction. To examine in vivo efficacy, EP was administered orally and Listeria monocytogenes given i.v. Mice were sacrificed three days post-infection to determine bacterial load in the spleen. We demonstrate that an endotoxin-free EP extract activates the innate immune response, stimulating production of IL-6, TNF, IL-12, and NO from macrophages in vitro. Along with evidence of enhanced macrophage function, we found that oral EP reduces bacterial burden during infection by Listeria monocytogenes, demonstrating its efficacy in vivo. EP initiates a signaling cascade within macrophages through both TLR4-dependent and -independent mechanisms, involving ERK, p38 and JNK, and ultimately the activation of NF-kappaB.


Assuntos
Echinacea , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Polissacarídeos/farmacologia , Administração Oral , Animais , Western Blotting , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Echinacea/química , Ensaio de Imunoadsorção Enzimática , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/isolamento & purificação , Lipopolissacarídeos/farmacologia , Listeriose/imunologia , Listeriose/prevenção & controle , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosforilação , Componentes Aéreos da Planta , Extratos Vegetais/farmacologia , Raízes de Plantas , Polimixina B/farmacologia , Polissacarídeos/administração & dosagem , Polissacarídeos/isolamento & purificação , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo
17.
Transpl Immunol ; 19(2): 103-11, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18503885

RESUMO

We have developed a model of aortic allograft vasculopathy (AV) that uses mouse strains that are fully disparate at Class I, Class II and minor histocompatibility antigens. Acute rejection is ablated with therapeutic doses of the calcineurin inhibitor Cyclosporine A (CyA). In this way we successfully mimic human disease. Using this model we have demonstrated, with cell transfer models using highly purified T cell populations, that calcineurin inhibitors ablate CD4(+) T cell effector mechanisms. As such, in the presence of calcineurin inhibition, graft vasculopathy is dependent on CD8(+) T cell effector mechanisms. In this study we examine the etiology of graft vasculopathy by these CD8(+) T cells in the presence of calcineurin inhibition. We transferred CD8(+) T cells from CyA treated IFN-gamma deficient mice into immunodeficient mouse recipients of aortic allografts to demonstrate that IFN-gamma production by CD8(+) T cells is essential for the development of AV in the presence of calcineurin inhibition. Using two models of CTL ablation we also demonstrated that CTL activity by CD8(+) T cells is essential for the development of AV in the presence of calcineurin inhibition. This is in contrast to models without calcineurin inhibitor immunosuppression where either pathway is capable, by itself, of inducing AV. These data indicate that although calcineurin inhibition ablates CD4(+) T cell effects and weakens CD8(+) T cell pathways, the antigenic challenge of the graft is enough to induce sufficient responsiveness from CD8(+) T cells to induce robust AV.


Assuntos
Aorta Abdominal/transplante , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Inibidores de Calcineurina , Interferon gama/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Aorta Abdominal/imunologia , Aorta Abdominal/patologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Ciclosporina/farmacologia , Imunossupressores/farmacologia , Interferon gama/imunologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Transplante Homólogo
18.
Transpl Immunol ; 19(2): 120-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18503887

RESUMO

The respective roles of the endothelium and the media as allo-immune targets in the generation of allograft vasculopathy (AV) have yet to be clearly defined. Although endothelial damage has been implicated in the progression of AV, evidence from mechanical vascular injury models suggests that medial injury may play a more dominant role. The overall objective of this research was to determine the relative importance of the endothelium versus the media as a target for immune injury and induction of AV. To investigate this we developed a novel model which involved the creation of chimeric aortic segments. To accomplish this we removed aortic segments from C3H/HeJ (C3H) mice and stripped them of endothelium by a short pulse with EDTA. The stripped C3H grafts were implanted into immunodeficient C57BL/6 (B6) RAG1(-/-) mice for a period of 21 days. As the immunodeficient mice did not mount an allo-immune response to the grafts, the endothelium was renewed by normal repair mechanisms. The new endothelium was recipient in origin, resulting in a chimeric graft with C3H media and B6 endothelium. We confirmed complete denudement by immunocytochemistry for endothelial specific markers, as well as by transmission and scanning electron microscopy. Replacement of endothelium with recipient endothelial cells was confirmed by immunocytochemistry, electron microscopy and by using a green fluorescent protein mouse transplant combination. Subsequent re-transplantation of the chimeric grafts into either B6 or C3H recipients demonstrated that an allogeneic media is more important than an allogeneic endothelium in inducing robust AV.


Assuntos
Aorta/imunologia , Aorta/transplante , Endotélio Vascular/imunologia , Túnica Média/imunologia , Animais , Aorta/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Quimeras de Transplante , Transplante Homólogo
19.
J Thorac Cardiovasc Surg ; 135(4): 777-83, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18374756

RESUMO

OBJECTIVE: The study objectives were to (1) assess the efficacy of N,O-carboxymethyl chitosan on postsurgical adhesion formation after cardiac surgery using a rabbit cardiac injury model and (2) explore the mechanism of action of N,O-carboxymethyl chitosan in the prevention of postsurgical adhesions using in vitro experimentation. METHODS: In the rabbit cardiac injury model, cardiac injury was generated by abrading the anterior surface of the heart with gauze and desiccated with oxygen. The rabbits were then either treated with N,O-carboxymethyl chitosan gel and solution on the injured surface or not treated. Fourteen days or 3 months after surgery, the severity and area of adhesion between the heart and sternum were evaluated. In the in vitro adherence assay, murine fibroblasts and macrophages were labeled with (3)H-thymidine and added to sterile tissue culture plates that had been precoated with N,O-carboxymethyl chitosan solution, culture medium, or hyaluronic acid. After incubation, the cells adherent to the coated plates were harvested and the levels of (3)H-thymidine were measured. RESULTS: Animals treated with N,O-carboxymethyl chitosan gel and solution showed significantly (P < .01) reduced severity and area of adhesion formation. Murine fibroblasts and macrophages did not adhere to N,O-carboxymethyl chitosan-coated tissue culture plates, even in the presence of serum. CONCLUSION: The application of N,O-carboxymethyl chitosan gel and solution significantly reduces the severity of postsurgical adhesion formation after cardiac surgery in the rabbit model. The inability of fibroblasts to adhere to N,O-carboxymethyl chitosan-coated surfaces suggests that N,O-carboxymethyl chitosan may act as a biophysical barrier.


Assuntos
Materiais Biocompatíveis/administração & dosagem , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Quitosana/administração & dosagem , Aderências Teciduais/prevenção & controle , Administração Tópica , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibroblastos , Géis , Macrófagos , Coelhos , Soluções , Aderências Teciduais/etiologia
20.
Transpl Int ; 21(7): 679-87, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18435686

RESUMO

The long term goal of immunological therapy for transplantation is to induce antigen specific unresponsiveness. One approach of significant current interest is the induction of T regulatory (Treg) cells that downregulate immune responses in an antigen specific manner. In this study, we examined the nature of the immunological regulation initiated by oral exposure to alloantigen. We previously demonstrated that feeding of allogeneic donor splenocytes significantly prolongs kidney allograft survival in rats. Purified CD8+ graft infiltrating cells (GIC), but not CD4+ GIC transfer graft prolongation to naïve animals demonstrating the presence of a CD8+ Treg population in the graft. In this study, we provide evidence that is consistent with a hypothesis that the CD8+ Treg generated by oral exposure to alloantigen is an IL-10 secreting, gammadelta TCR+ T cell.


Assuntos
Transferência Adotiva , Linfócitos T CD8-Positivos/imunologia , Sobrevivência de Enxerto/imunologia , Isoantígenos/imunologia , Transplante de Rim , Receptores de Antígenos de Linfócitos T gama-delta/análise , Linfócitos T Reguladores/imunologia , Administração Oral , Animais , Linfócitos T CD8-Positivos/metabolismo , Isoantígenos/administração & dosagem , Masculino , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Baço/citologia , Baço/imunologia , Linfócitos T Reguladores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...