Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
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 ; 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
3.
Am J Pathol ; 183(2): 459-69, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23731726

RESUMO

Fibroblast progenitor cells (fibrocytes) are important to the development of myocardial fibrosis and are suggested to migrate to the heart via CXCL12 and chemokine ligand (CCL) 2. We hypothesized that if these chemokines are recruiting fibrocytes, disrupting their signaling will reduce early (3-day) fibrocyte infiltration and, consequently, fibrosis in the myocardium. C57/Bl6 and CCR2(-/-) mice were infused with saline or angiotensin (Ang) II, with or without CXC receptor 4 blockade (AMD3100). Hearts were assessed for chemokine up-regulation, immunofluorescence, and histological features. AngII caused early myocardial up-regulation of CXCL12 and CCL2, which corresponded to significant myocardial infiltration and fibrosis compared with controls. Animals receiving AMD3100 and/or with the genotype CCR2(-/-) failed to demonstrate reductions in infiltrate or fibrosis after 3 days of AngII, and AngII + AMD3100 animals showed exacerbated fibrocyte infiltration and fibrosis compared with AngII alone. CCR2(-/-) mice demonstrated significant reductions in myocardial fibrosis relative to wild type, but this was after 28 days of AngII infusion and was the result of reduced infiltrating cell proliferation. An alternative CCR2 ligand, CCL12, was found to be increasing infiltrating cell proliferation in the heart after AngII infusion, which we confirmed in vitro. In conclusion, early fibrocyte recruitment cannot be inhibited through modulating CXCL12 or CCL2, as previously thought. Ablating CCR2 signaling did confer myocardial fibrosis reductions, but these benefits were not observed until much later and were likely the result of modulated proliferation through ablating the CCL12-CCR2 interaction.


Assuntos
Angiotensina II/farmacologia , Quimiocina CCL2/fisiologia , Quimiocina CXCL12/fisiologia , Fibroblastos/fisiologia , Miocárdio/metabolismo , Vasoconstritores/farmacologia , Animais , Benzilaminas , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Ciclamos , Fibrose/patologia , Compostos Heterocíclicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Receptores CXCR4/antagonistas & inibidores , Células-Tronco/fisiologia , Regulação para Cima
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.
Artigo em Inglês | MEDLINE | ID: mdl-24721281

RESUMO

BACKGROUND: Myocardial fibrosis is a pathological process that is characterized by disrupted regulation of extracellular matrix proteins resulting in permanent scarring of the heart tissue and eventual diastolic heart failure. Pro-fibrotic molecules including transforming growth factor-ß and connective tissue growth factor are expressed early in the AngiotensinII (AngII)-induced and other models of myocardial fibrosis. As such, antibody-based therapies against these and other targets are currently under development. RESULTS: In the present study, C57Bl/6 mice were subcutaneously implanted with a mini-osmotic pump containing either AngII (2.0 µg/kg/min) or saline control for 3 days in combination with mIgG (1 mg/kg/d) injected through the tail vein. Fibrosis was assessed after picosirius red staining of myocardial cross-sections and was significantly increased after AngII exposure compared to saline control (11.37 ± 1.41%, 4.94 ± 1.15%; P <0.05). Non-specific mIgG treatment (1 mg/kg/d) significantly increased the amount of fibrosis (26.34 ± 3.03%; P <0.01). However, when AngII exposed animals were treated with a Fab fragment of the mIgG or mIgM, this exacerbation of fibrosis was no longer observed (14.49 ± 2.23%; not significantly different from AngII alone). CONCLUSIONS: These data suggest that myocardial fibrosis was increased by the addition of exogenous non-specific antibodies in an Fc-mediated manner. These findings could have substantial impact on the future experimental design of antibody-based therapeutics.

7.
Hypertens Res ; 35(4): 449-56, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22258030

RESUMO

Activation of the renin-angiotensin system (RAS) is thought to promote myocardial fibrosis. However, it is unclear whether this physiological fibrotic response results from chronic hemodynamic stress or from direct cellular signaling. Male C57B/6 mice were randomly assigned to receive angiotensin II (AngII) (2.0 µg kg(-1) min(-1)), AngII+hydralazine (6.9 µg kg(-1) min(-1)) or saline (control) via osmotic pumps for 7 days. Blood pressure was measured via noninvasive plethysmography. Hearts were harvested and processed for analysis. Cellular infiltration and collagen deposition were analyzed using histological staining. Molecular mediators were assessed using quantitative RT-PCR. As previously described, animals that received AngII developed hypertension and multifocal cellular infiltration by SMA(+)/CD133(+) fibroblast progenitors followed by collagen deposition. The coadministration of hydralazine with AngII completely inhibited the hypertensive effects of AngII (P0.01) and resulted in minimal cellular infiltration and minimal collagen deposition. These findings were in the context of persistent RAS activation, which was evidenced by elevation in serum aldosterone levels in animals that received AngII or AngII+hydralazine compared with animals that received saline. At the molecular level, infusion of AngII resulted in the significant upregulation of profibrotic factors (connective tissue growth factor-7.8±0.7 fold), proinflammatory mediators (TNFα-4.6±0.8 fold; IL-1ß-6.4±2.6 fold) and chemokines (CCL2-3.8±1.0 fold; CXCL12-3.2±0.4 fold), which were inhibited when hydralazine was also infused. We provide evidence that myocardial infiltration by fibroblast progenitor cells secondary to AngII and the resultant fibrosis can be prevented by the addition of hydralazine. Furthermore, the beneficial effects of hydralazine were observed while maintaining RAS activation, suggesting that the mechanism of fibrosis is blood pressure dependent.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/fisiologia , Movimento Celular/fisiologia , Fibroblastos/patologia , Hipertensão/patologia , Miocárdio/patologia , Aldosterona/sangue , Animais , Pressão Sanguínea/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Colágeno/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Coração/efeitos dos fármacos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Masculino , Camundongos , Miocárdio/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia
8.
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
9.
J Renin Angiotensin Aldosterone Syst ; 12(3): 295-303, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21393356

RESUMO

HYPOTHESIS: Monocytes produce pro-inflammatory cytokines in response to Angiotensin II (AngII). INTRODUCTION: AngII has been suggested by many to be pro-inflammatory and likely to contribute to the migration of leukocytes in patients with cardiovascular conditions. MATERIALS AND METHODS: Monocytes were purified from peripheral blood mononuclear cells (PBMCs) by negative selection using antibodies conjugated to magnetic beads. Detection of CD14(+) and AT(1)R expression was achieved by double-labeling flow cytometry. Highly purified monocytes were then stimulated with AngII (6 and 24 h) to assess IL-6 and TNF-α transcript levels by qRT-PCR and protein secretion by ELISA. RESULTS: Monocytes comprised 9.7 ± 2.0% of the PBMCs. Monocyte isolation by negative selection yielded a purity of up to 99.8%. We demonstrated AT(1)R expression on 9.5 ± 0.3% of highly purifed CD14(+)/CD16(-) monocytes. Stimulation of highly purified monocytes with AngII resulted in increased transcript levels of IL-6 at 6 h but not at 24 h, and increased secretion of IL-6 in a dose-dependent manner compared with controls (p <0.01). Conversely, there was no increase in TNF-α mRNA transcripts or protein secretion. CONCLUSIONS: We provide evidence that a CD14(+)/CD16(-) subset of highly purified human monocytes express AT(1)R and respond to AngII exposure in vitro by producing IL-6 but not TNF-α.


Assuntos
Angiotensina II/farmacologia , Interleucina-6/biossíntese , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Adulto , Separação Celular , Feminino , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-6/genética , Receptores de Lipopolissacarídeos/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de IgG/metabolismo , Fator de Necrose Tumoral alfa/genética
10.
J Invest Surg ; 23(2): 79-86, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20497009

RESUMO

AIMS: Characterize mononuclear cell migration after acute-myocardial infarction (MI). MATERIAL AND METHODS: Male Lewis rats underwent a left thoracotomy and ligation of the left anterior descending coronary artery (MI group). Control animals underwent thoracotomy without ligation (Sham group). Animals were sacrificed at 0, 2, 4, or 24 hr after the onset of ischemia. Leukocyte migration was assessed using isolated and In(111) labeled mononuclear cells (injected at the onset of ischemia) and gamma-count determined at 24 hours. Inhibition of migration was evaluated with monoclonal anti alpha4 and/or beta2 antibodies. RESULTS: Serum troponin was significantly elevated in animals with MI as compared with Sham (p = .017). Labeled mononuclear cell migration was five-fold higher in MI-treated animals than in Sham (p = .006). ED-1 positive mononuclear cells were confirmed in the left myocardium after 24 hr of ischemia. MCP-1 mRNA was significantly elevated in the left myocardium at 2 hr and 4 hr and peaked at 24 hr (p <.05). In addition, alpha4 integrin blockade inhibited labeled mononuclear cell migration by 22%. Blockade of beta2 integrin inhibited mononuclear cell migration by 48%, while the combined alpha4+beta2 blockade resulted in 59% inhibition of labeled mononuclear cell migration compared with treatment with isotype control antibody (p = .001). CONCLUSIONS: Significant ED1+ mononuclear cell migration within 24 hr of MI correlated with peak MCP-1 mRNA. Monoclonal antibody blockade suggested that early mononuclear cell migration is dependent only in part on alpha4 and beta2 integrins.


Assuntos
Antígenos CD18/imunologia , Movimento Celular/imunologia , Integrina alfa4/imunologia , Monócitos/fisiologia , Infarto do Miocárdio/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Quimiocina CCL2/genética , Masculino , Monócitos/imunologia , Miocárdio/imunologia , Ratos , Ratos Endogâmicos Lew , Troponina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA