Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(4): e0176147, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28448522

RESUMO

Renal and lung fibrosis was characterized by the accumulation of collagen-immunoreactive mesenchymal cells expressing the intermediate filament protein nestin. The present study tested the hypothesis that nestin expression was increased in the hypertrophied/fibrotic left ventricle of suprarenal abdominal aorta constricted adult male Sprague-Dawley rats and induced in ventricular fibroblasts by pro-fibrotic peptide growth factors. Nestin protein levels were upregulated in the pressure-overloaded left ventricle and expression positively correlated with the rise of mean arterial pressure. In sham and pressure-overloaded hearts, nestin immunoreactivity was detected in collagen type I(+)-and CD31(+)-cells identified in the interstitium and perivascular region whereas staining was absent in smooth muscle α-actin(+)-cells. A significantly greater number of collagen type I(+)-cells co-expressing nestin was identified in the left ventricle of pressure-overloaded rats. Moreover, an accumulation of nestin(+)-cells lacking collagen, CD31 and smooth muscle α-actin staining was selectively observed at the adventitial region of predominantly large calibre blood vessels in the hypertrophied/fibrotic left ventricle. Angiotensin II and TGF-ß1 stimulation of ventricular fibroblasts increased nestin protein levels via phosphatidylinositol 3-kinase- and protein kinase C/SMAD3-dependent pathways, respectively. CD31/eNOS(+)-rat cardiac microvascular endothelial cells synthesized/secreted collagen type I, expressed prolyl 4-hydroxylase and TGF-ß1 induced nestin expression. The selective accumulation of adventitial nestin(+)-cells highlighted a novel feature of large vessel remodelling in the pressure-overloaded heart and increased appearance of collagen type I/nestin(+)-cells may reflect an activated phenotype of ventricular fibroblasts. CD31/collagen/nestin(+)-interstitial cells could represent displaced endothelial cells displaying an unmasked mesenchymal phenotype, albeit contribution to the reactive fibrotic response of the pressure-overloaded heart remains unknown.


Assuntos
Colágeno/metabolismo , Mesoderma/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Nestina/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Regulação para Cima , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/fisiopatologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fibrose , Hipertrofia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Mesoderma/efeitos dos fármacos , Microvasos/patologia , Contração Miocárdica/efeitos dos fármacos , Fenótipo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
2.
J Cell Physiol ; 232(7): 1717-1727, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27439108

RESUMO

Nestin(+) -cardiomyocytes were identified in the ischemically damaged human/rodent heart, albeit the cellular source, and signaling events implicated in the appearance of the intermediate filament protein remained undefined. Expression of the enhanced green fluorescent protein (EGFP) driven by the second intron of the nestin gene identified a subpopulation of EGFP/nestin(+) cells that differentiated to a vascular phenotype in the peri-infarct/infarct region of post-MI mice albeit the transgene was not detected in nestin(+) -cardiomyocytes. α-MHC-driven expression of the reporter mCherry was detected in troponin-T(+) - and nestin(+) -cardiomyocytes in the peri-infarct/infarct region of post-MI mice. However, the cell cycle re-entry of nestin/mCherry(+) -cardiomyocytes was not observed. Nestin staining was identified in a paucity of neonatal rat ventricular cardiomyocytes (NNVM). Exposure to phorbol 12,13-dibutyrate (PDBu) induced NNVM hypertrophy but did not promote nestin expression or Brdu incorporation. PDBu treatment of NNVMs phosphorylated p38 MAPK and HSP27 and HSP27 phosphorylation was abrogated by the p38 MAPK inhibitor SB203580. PDBu/SB203580 co-treatment significantly increased the percentage of NNVMs that expressed nestin and incorporated Brdu. In the heart of embryonic 10.5 day mice, nestin immunoreactivity was observed in cycling troponin-T(+) -cardiomyocytes. Nestin was also detected in embryonic rat ventricular cardiomyocytes and depletion of the intermediate filament protein attenuated cell cycle re-entry. Thus, nestin expressed by pre-existing cardiomyocytes following ischemic damage recapitulated in part an embryonic trait and may provide the requisite phenotype to initiate cell cycle re-entry. However, the overt activation of the p38 MAPK pathway post-MI may in part limit the appearance and inhibit the cell cycle re-entry of nestin(+) -cardiomyocytes. J. Cell. Physiol. 232: 1717-1727, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Embrião de Mamíferos/citologia , Miócitos Cardíacos/enzimologia , Nestina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Ciclo Celular , Elementos Facilitadores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Ventrículos do Coração/patologia , Íntrons/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Acetato de Tetradecanoilforbol/farmacologia , Transgenes
3.
J Cell Physiol ; 230(1): 170-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24915827

RESUMO

Upregulation of the intermediate filament protein nestin was identified in a subpopulation of fibroblasts during reactive and reparative fibrosis and directly contributed to the enhanced proliferative phenotype. The present study tested the hypothesis that nestin was expressed in lung fibroblasts and the pattern of expression represented a distinct marker of pulmonary remodeling secondary to myocardial infarction and type I diabetes. Nestin((+)) fibroblasts were detected in rat lungs and a subpopulation exhibited a myofibroblast phenotype delineated by the co-expression of smooth muscle α-actin. In the lungs of myocardial infarcted rats, interstitial collagen content and nestin mRNA/protein levels were significantly increased despite the absence of secondary pulmonary hypertension, whereas smooth muscle α-actin protein expression was unchanged. Exposure of rat pulmonary fibroblasts to pro-fibrotic stimuli angiotensin II and transforming growth factor-ß significantly increased nestin protein levels. In the lungs of type I diabetic rats, the absence of a reactive fibrotic response was associated with a significant downregulation of nestin mRNA/protein expression. Nestin was reported a target of miR-125b, albeit miR-125b levels were unchanged in pulmonary fibroblasts treated with pro-fibrotic stimuli. Nestin((+)) cells lacking smooth muscle α-actin/collagen staining were also identified in rodent lungs and a transgenic approach revealed that expression of the intermediate filament protein was driven by intron 2 of the nestin gene. The disparate regulation of nestin characterized a distinct pattern of pulmonary remodeling secondary to myocardial infarction and type I diabetes and upregulation of the intermediate filament protein in lung fibroblasts may have facilitated in part the reactive fibrotic response.


Assuntos
Remodelação das Vias Aéreas , Diabetes Mellitus Tipo 1/patologia , Pulmão/patologia , Infarto do Miocárdio/patologia , Nestina/biossíntese , Actinas/biossíntese , Angiotensina II/farmacologia , Animais , Biomarcadores , Diferenciação Celular , Colágeno Tipo I/biossíntese , Fibroblastos/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Hipertensão Pulmonar/patologia , Hipertrofia Ventricular Direita/patologia , Pulmão/metabolismo , Masculino , MicroRNAs/biossíntese , MicroRNAs/genética , Contração Miocárdica/fisiologia , Nestina/genética , Fibrose Pulmonar/patologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Estreptozocina , Fator de Crescimento Transformador beta/farmacologia
4.
Cardiovasc Diabetol ; 12: 114, 2013 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-23938193

RESUMO

BACKGROUND: Cardiac nestin(+) cells exhibit properties of a neural progenitor/stem cell population characterized by the de novo synthesis of neurofilament-M in response to ischemic injury and 6-hydroxydopamine administration. The induction of growth associated protein 43 (GAP43) was identified as an early event of neurogenesis. The present study tested the hypothesis that the de novo synthesis of neurofilament-M by nestin(+) cells was preceded by the transient upregulation of GAP43 during the acute phase of reparative fibrosis in the infarcted male rat heart. Secondly, a seminal feature of diabetes is impaired wound healing secondary to an inadequate neurogenic response. In this regard, an additional series of experiments tested the hypothesis that the neurogenic response of cardiac nestin(+) cells was attenuated in a setting of type I diabetes. METHODS: The neurogenic response of cardiac nestin(+) cells was examined during the early phase of reparative fibrosis following permanent ligation of the left anterior descending coronary artery in the adult male rat heart. The experimental model of type I diabetes was created following a single injection of streptozotocin in adult male rats. The impact of a type I diabetic environment on the neurogenic response of cardiac nestin(+) cells was examined during myocardial infarction and following the administration of 6-hydroxydopamine. RESULTS: During the early phase of scar formation/healing, the density of GAP43/nestin(+) fibres innervating the peri-infarct/infarct region was significantly increased, whereas neurofilament-M/nestin(+) fibres were absent. With ongoing scar formation/healing, a temporal decrease of GAP43/nestin(+) fibre density and a concomitant increase in the density of innervating neurofilament-M/nestin(+) fibres were observed. The neurogenic response of cardiac nestin(+) cells during scar formation/healing was inhibited following the superimposition of type I diabetes. The de novo synthesis of neurofilament-M by nestin(+) cells after 6-hydroxydopamine administration was likewise attenuated in the heart of type I diabetic rats whereas the density of GAP43/nestin(+) fibres remained elevated. CONCLUSION: The transient upregulation of GAP43 apparently represents a transition event during the acquisition of a neuronal-like phenotype and a type I diabetic environment attenuated the neurogenic response of cardiac nestin(+) cells to ischemia and 6-hydroxydopamine.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Proteína GAP-43/metabolismo , Coração/inervação , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Nestina/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Animais , Cicatriz/metabolismo , Cicatriz/patologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Fibrose , Masculino , Infarto do Miocárdio/patologia , Miocárdio/patologia , Fibras Nervosas/metabolismo , Fibras Nervosas/patologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Proteínas de Neurofilamentos/metabolismo , Neurogênese/efeitos dos fármacos , Oxidopamina/farmacologia , Fenótipo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...