Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Ethnopharmacol ; 106(3): 360-3, 2006 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-16504434

RESUMO

The root of Heracleum maximum Bartr. (Umbelliferae), known to possess direct antifungal and anti-mycobacterial properties, has been reported anecdotally to possess antiviral properties. It was therefore hypothesized that the plant may have immunostimulant properties. This hypothesis was tested using a macrophage activation assay to evaluate the ability of aqueous extracts of the root of Heracleum maximum to stimulate IL-6 production. All Heracleum maximum extracts were found to stimulate IL-6 and produced a steep dose-response curve. With the assay performed twice in the absence of the macrophage primer, IFN-gamma, the mean IL-6 production in the setting of the strongest extract was 3648pg/ml (95% CI 3361-3935) and 5430pg/ml (95% CI 4976-5885) as compared to 2722pg/ml (95% CI 2620-2824) and 6772pg/ml (95% CI 6282-7262) produced by the LPS positive control, respectively. In the presence of IFN-gamma, the strongest extract produced a mean concentration of IL-6 of 21804pg/ml (95% CI 19755-23854) surpassing the 14893pg/ml (13159-16628) produced by the LPS+IFN-gamma positive control. These positive results confirm the hypothesis of immunostimulation and thus support the anecdotal reports of antiviral activity.


Assuntos
Adjuvantes Imunológicos/farmacologia , Heracleum , Interleucina-6/biossíntese , Macrófagos/efeitos dos fármacos , Fitoterapia , Extratos Vegetais/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Macrófagos/metabolismo , Camundongos , Extratos Vegetais/administração & dosagem , Extratos Vegetais/uso terapêutico , Raízes de Plantas
11.
Cardiovasc Res ; 65(1): 283-91, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15621057

RESUMO

OBJECTIVE: Allograft vasculopathy (AV) has emerged as the major obstacle to long-term survival in clinical heart transplantation. Immune events are implicated in the development of AV, but the cellular and molecular mechanisms involved remain unclear. We sought to determine whether and by what mechanism CD8(+) T lymphocytes are able to generate AV in a murine aortic allograft model. METHODS: Allo-primed CD8(+) T lymphocytes were transferred into immunodeficient (RAG-1(-/-)) mouse recipients of aortic allografts. We also transferred primed CD8(+) T cells with targeted deletions of effector molecules (perforin, Fas-ligand) to determine the role of direct cytolysis (CTL) in CD8(+) T-cell-mediated AV. We determined the role of non-CTL effector mechanisms through the transfer of either wildtype or interferon-gamma (IFN-gamma)-deficient CD8(+) T cells into RAG-1(-/-) recipients of MHC class I-deficient allografts. RESULTS: Adoptive transfer of primed wildtype CD8(+) T lymphocytes into immunodeficient recipients of aortic allografts resulted in the development of robust AV lesions. Transfer of CD8(+) T lymphocytes with targeted deletions in CTL effector molecules resulted in reduction of AV lesion size but not abrogation. Transfer of wildtype CD8(+) T cells into recipients of MHC class I-deficient grafts resulted in a reduction in AV lesion size, while transfer of interferon-gamma-deficient CD8(+) T cells into MHC class I-deficient grafts abrogated AV. CONCLUSIONS: These data indicate that CD8(+) T cells mediate AV through direct cytolysis and a distinct interferon-gamma-dependent non-CTL effector pathway. Given the resistance of this cell type to conventional immunosuppression, these results may have important therapeutic implications.


Assuntos
Aorta Abdominal/transplante , Linfócitos T CD8-Positivos/imunologia , Interferon gama/imunologia , Neovascularização Patológica , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/transplante , Morte Celular , Ciclosporina/farmacologia , Testes Imunológicos de Citotoxicidade , Proteína Ligante Fas , Deleção de Genes , Imunização , Imunossupressores/farmacologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Perforina , Proteínas Citotóxicas Formadoras de Poros , Imunologia de Transplantes , Transplante Homólogo
12.
Transpl Immunol ; 15(1): 35-44, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16223671

RESUMO

We investigated the role of CD4(+) T cells and CD8(+) T cells in mediating allograft vasculopathy in Cyclosporin A (CyA) immunosuppressed mice. We first established that a dose of 50 mg/kg/d CyA was required to prevent acute rejection in C57BL/6 mice. CyA given at 50 mg/kg/d did not prevent allograft vasculopathy in either cardiac or aortic transplants in these mice. Using CD4(-/-) and CD8(-/-) mice, we established that CyA immunosuppression at this dose was only effective at preventing allograft vasculopathy in mice lacking CD8(+) T cells. This implicates CD8(+) T cells in the development of AV in situations of clinical cardiac transplantation where CyA is still the mainstay of immunosuppressive therapy. We confirmed the important role for CD8(+) T cells in AV in the face of CyA immunosuppression by allopriming mice in the presence of CyA and transferring alloprimed T cells into RAG1(-/-) immunodeficient mice. The RAG1(-/-) mice were also treated with CyA. In this situation (CyA present during the allopriming and in the recipient), only primed CD8(+) T cells could mediate AV, primed CD4(+) T cells could not. Alloprimed CD8(+) T cells raised in the presence of CyA exhibited markedly reduced direct recognition responses (as measured by MLR) and effector responses (as measured by cytotoxic activity). In contrast indirect activation was retained. We interpret these data to suggest that in the face of CyA immunosuppression CD4(+) T cell effector function is ablated while CD8(+) T cell function remains partially intact. The in vitro data suggest that the indirect pathway remains intact in this population of CyA resistant CD8(+) T cells.


Assuntos
Doenças da Aorta/imunologia , Linfócitos T CD8-Positivos/imunologia , Ciclosporina/farmacologia , Transplante de Coração/imunologia , Imunossupressores/farmacologia , Animais , Aorta/imunologia , Aorta/patologia , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Ciclosporina/uso terapêutico , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica , Terapia de Imunossupressão , Imunossupressores/uso terapêutico , Camundongos
13.
Transpl Immunol ; 14(1): 27-35, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15814279

RESUMO

We investigated the role of CD4+ and CD8+ T subsets as well as T cell cytolytic effector mechanisms in the aortic allograft model of allograft vasculopathy using CD4 and CD8 gene knockout mice (CD4(-/-), CD8(-/-)) and mice deficient in cytolytic effector pathways. Medial apoptosis at 2 weeks was reduced in CD8(-/-) mice and in mice where cytotoxic T cell activity was compromised. At 8 weeks, substantial medial damage was observed in wild-type (WT) and CD4(-/-) recipients but medial preservation was evident in CD8(-/-) mice and in mice with impaired cytotoxic T cell activity. The intima/media ratio, a comprehensive measure of allograft vasculopathy, was similar in WT and CD4(-/-) recipients but was significantly reduced in CD8(-/-) mice and mice with impaired cytotoxic T cell activity. These data indicate that CD8+ T cells contribute to the vascular remodeling that is characteristic of allograft vasculopathy. They also show that CD8+ T cells participate in allograft vasculopathy in the absence of CD4+ T cell help. We further demonstrated that WT mice exhibited robust allograft vasculopathy in the presence of cyclosporin A immunosuppression but that allograft vasculopathy was ablated in cyclosporin-treated CD8(-/-) mice. This supports the hypothesis that non-CD8+ T cell effector mechanisms are sensitive to calcineurin inhibitor therapy but that CD8+ T cell-mediated allograft vasculopathy is refractory to such treatment. Taken together, our data suggest that CD8+ T cells contribute to the induction of vascular remodeling in allograft vasculopathy and provide evidence that novel therapies which target CD8+ T cell effector function might be effective in mitigating AV in the clinical setting.


Assuntos
Aorta/transplante , Linfócitos T CD4-Positivos/imunologia , Rejeição de Enxerto/imunologia , Músculo Liso Vascular/patologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose , Rejeição de Enxerto/patologia , Masculino , Camundongos , Camundongos Knockout , Transplante Homólogo
14.
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
15.
Int J Oncol ; 25(6): 1693-9, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15547707

RESUMO

Immunotherapy based on the administration of the mycobacterium bacillus Calmette-Guerin has been successfully used in the treatment of in situ transitional cell bladder cancer, and may be applicable to the treatment of cutaneous malignant melanoma. Antigen 85A (Ag85A) and heat shock protein 65 kDa (hsp65) are major secreted proteins of Mycobacterium species and potent stimulators of cell-mediated immunity. This study evaluated the ability of Ag85A and hsp65 gene transfection to limit tumor growth by B16-F10 mouse melanoma cells. Immunoblotting confirmed protein expression and secretion by B16-F10 cells that were transiently transfected with plasmid DNA containing the Ag85A or hsp65 gene. Groups of syngeneic C57BL/6 mice were injected subcutaneously with 1x10(5) untransfected B16-F10 cells or B16-F10 cells transiently transfected with either empty vector or vector containing the Ag85A or hsp65 gene. Ag85A-expressing B16-F10 cells exhibited a dramatic 76% reduction (p<0.05, Mann-Whitney U test) in tumor weight in comparison to empty vector controls at 14 days post-inoculation. In contrast, hsp65-transfected B16-F10 cells did not show any change in tumorigenicity. Decreased tumorigenicity by Ag85A-transfected B16-F10 cells was not due to a reduced ability of Ag85A-transfected B16-F10 cells to proliferate since both mock- and Ag85A-transfected B16-F10 cells showed increased in vitro proliferation in comparison to untransfected cells. Hematoxylin and eosin staining revealed that Ag85A-transfected B16-F10 tumors contained an inflammatory leukocyte infiltrate that was not present in hsp65-transfected tumors. Reduced tumor progression by Ag85A-transfected B16-F10 melanoma cells suggests that immunotherapy based on the transient induction of Ag85A expression may be an effective approach for the treatment of cutaneous malignant melanoma.


Assuntos
Antígenos de Bactérias/imunologia , Melanoma/genética , Melanoma/imunologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Animais , Vacina BCG/uso terapêutico , Proliferação de Células , Progressão da Doença , Feminino , Humanos , Imunoterapia , Melanoma/veterinária , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium bovis , Neoplasias Experimentais , Neoplasias Cutâneas/veterinária , Transfecção
16.
J Heart Lung Transplant ; 22(3): 357-60, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12633704

RESUMO

To determine the nature of the proliferative lesion in obliterative airway disease, heterotopic tracheal allograft transplantation was performed between fully disparate Brown Norway and Lewis rat strains. Four weeks after transplantation, the resulting lumenal occlusive lesion was stripped from the underlying tissue. The lesion was probed using immunohistochemical analysis with monoclonal antibodies and for DNA using strain-specific primers for Brown Norway or Lewis major histocompatibility complex Class I alleles. The lesions were alpha-actin positive, and polymerase chain reaction probing revealed only recipient DNA in the lesion tissue, regardless of the direction of transplantation, with no amplification of donor DNA. From this, we conclude that the proliferative lesion in the rat heterotopic tracheal model is of recipient origin a finding with important implications for the pathobiology of obliterative airway disease.


Assuntos
Bronquiolite Obliterante/patologia , Traqueia/transplante , Transplante Heterotópico , Animais , Masculino , Omento , Reação em Cadeia da Polimerase , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Transplante Homólogo
17.
Surgery ; 135(3): 307-12, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14976481

RESUMO

BACKGROUND: Postoperative adhesions have proven to be intractable complications after abdominal operations. This study assessed the efficacy of N, O - carboxymethyl chitosan (NOCC) to limit adhesion formation and re-formation in a rabbit abdominal surgery model. METHODS: In study 1 (adhesion formation), injuries to the large bowel, cecum, and abdominal sidewall were generated in rabbits. The rabbits (10/group) were randomly assigned to 1 of 5 treatment groups: Group A received no NOCC treatment; in group B, NOCC gel was applied directly to the injured site and NOCC solution was applied throughout the abdominal cavity; in group C, NOCC gel was applied near the injured site and NOCC solution was applied as above; in group D, NOCC gel was applied distant to the injury and NOCC solution was applied as above; in group E, a mixture of NOCC gel and solution was applied at the injured site. Adhesions were evaluated 14 days later. In study 2 (adhesion re-formation), adhesions were generated as above but were then lysed by careful dissection. After adhesiolysis, the rabbits (9/group) were treated with NOCC gel and solution at the site of adhesiolysis or left untreated. Adhesion re-formation was assessed 14 days later. In study 3 (mechanism of action), sterile tissue culture plates were coated with NOCC and adhesion of cultured, radiolabeled murine fibroblasts to the plates was assessed. RESULTS: In study 1, animals treated with NOCC gel and solution showed reduced adhesion formation (P<.01). NOCC gel was equally efficacious if applied on the site of injury or near the site of injury but less efficacious if applied at a site distant to the injury. In study 2, animals treated with NOCC gel and solution showed less adhesion re-formation compared with the untreated control animals (P<.01). In study 3, murine fibroblasts did not adhere to NOCC-coated tissue culture plates. CONCLUSIONS: NOCC gel and solution can reduce adhesion formation and re-formation in this rabbit model. The inability of fibroblasts to adhere to NOCC solution-coated surfaces suggests that NOCC may act as a biophysical barrier.


Assuntos
Materiais Biocompatíveis/administração & dosagem , Quitina/análogos & derivados , Quitina/administração & dosagem , Quitosana , Géis/administração & dosagem , Laparotomia/efeitos adversos , Soluções/administração & dosagem , Aderências Teciduais/prevenção & controle , Músculos Abdominais/lesões , Parede Abdominal/fisiopatologia , Administração Tópica , Animais , Colo/lesões , Colo/fisiopatologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Técnicas In Vitro , Camundongos , Modelos Animais , Peritônio/lesões , Coelhos , Recidiva , Aderências Teciduais/etiologia
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA