Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Am J Physiol Renal Physiol ; 318(2): F285-F297, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31760770

RESUMO

Juxtaglomerular (JG) cells, major sources of renin, differentiate from metanephric mesenchymal cells that give rise to JG cells or a subset of smooth muscle cells of the renal afferent arteriole. During periods of dehydration and salt deprivation, renal mesenchymal stromal cells (MSCs) differentiate from JG cells. JG cells undergo expansion and smooth muscle cells redifferentiate to express renin along the afferent arteriole. Gene expression profiling comparing resident renal MSCs with JG cells indicates that the transcription factor Sox6 is highly expressed in JG cells in the adult kidney. In vitro, loss of Sox6 expression reduces differentiation of renal MSCs to renin-producing cells. In vivo, Sox6 expression is upregulated after a low-Na+ diet and furosemide. Importantly, knockout of Sox6 in Ren1d+ cells halts the increase in renin-expressing cells normally seen during a low-Na+ diet and furosemide as well as the typical increase in renin. Furthermore, Sox6 ablation in renin-expressing cells halts the recruitment of smooth muscle cells along the afferent arteriole, which normally express renin under these conditions. These results support a previously undefined role for Sox6 in renin expression.


Assuntos
Arteríolas/metabolismo , Sistema Justaglomerular/irrigação sanguínea , Células-Tronco Mesenquimais/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Renina/metabolismo , Fatores de Transcrição SOXD/metabolismo , Animais , Arteríolas/efeitos dos fármacos , Pressão Sanguínea , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Dieta Hipossódica , Diuréticos/farmacologia , Furosemida/farmacologia , Regulação da Expressão Gênica , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Renina/genética , Fatores de Transcrição SOXD/deficiência , Fatores de Transcrição SOXD/genética , Transdução de Sinais
2.
Circ Res ; 120(9): 1403-1413, 2017 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-28209718

RESUMO

RATIONALE: Direct reprogramming of cardiac fibroblasts to cardiomyocytes has recently emerged as a novel and promising approach to regenerate the injured myocardium. We have previously demonstrated the feasibility of this approach in vitro and in vivo using a combination of 4 microRNAs (miR-1, miR-133, miR-208, and miR-499) that we named miR combo. However, the mechanism of miR combo mediated direct cardiac reprogramming is currently unknown. OBJECTIVE: Here, we investigated the possibility that miR combo initiated direct cardiac reprogramming through an epigenetic mechanism. METHODS AND RESULTS: Using a quantitative polymerase chain reaction array, we found that histone methyltransferases and demethylases that regulate the trimethylation of H3K27 (H3K27me3), an epigenetic modification that marks transcriptional repression, were changed in miR combo-treated fibroblasts. Accordingly, global H3K27me3 levels were downregulated by miR combo treatment. In particular, the promoter region of cardiac transcription factors showed decreased H3K27me3 as revealed by chromatin immunoprecipitation coupled with quantitative polymerase chain reaction. Inhibition of H3K27 methyltransferases or of the PRC2 (Polycomb Repressive Complex 2) by pharmaceutical inhibition or siRNA reduced the levels of H3K27me3 and induced cardiogenic markers at the RNA and protein level, similarly to miR combo treatment. In contrast, knockdown of the H3K27 demethylases Kdm6A and Kdm6B restored the levels of H3K27me3 and blocked the induction of cardiac gene expression in miR combo-treated fibroblasts. CONCLUSIONS: In summary, we demonstrated that removal of the repressive mark H3K27me3 is essential for the induction of cardiac reprogramming by miR combo. Our data not only highlight the importance of regulating the epigenetic landscape during cell fate conversion but also provide a framework to improve this technique.


Assuntos
Reprogramação Celular , Metilação de DNA , Epigênese Genética , Fibroblastos/metabolismo , Histonas/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Reprogramação Celular/efeitos dos fármacos , Técnicas de Reprogramação Celular , Metilação de DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica , Células HEK293 , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histonas/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Metilação , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Miócitos Cardíacos/efeitos dos fármacos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Interferência de RNA , Transdução de Sinais , Transfecção
3.
Circ Res ; 116(10): 1700-11, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25953925

RESUMO

The human heart has a limited capacity to regenerate lost or damaged cardiomyocytes after cardiac insult. Instead, myocardial injury is characterized by extensive cardiac remodeling by fibroblasts, resulting in the eventual deterioration of cardiac structure and function. Cardiac function would be improved if these fibroblasts could be converted into cardiomyocytes. MicroRNAs (miRNAs), small noncoding RNAs that promote mRNA degradation and inhibit mRNA translation, have been shown to be important in cardiac development. Using this information, various researchers have used miRNAs to promote the formation of cardiomyocytes through several approaches. Several miRNAs acting in combination promote the direct conversion of cardiac fibroblasts into cardiomyocytes. Moreover, several miRNAs have been identified that aid the formation of inducible pluripotent stem cells and miRNAs also induce these cells to adopt a cardiac fate. MiRNAs have also been implicated in resident cardiac progenitor cell differentiation. In this review, we discuss the current literature as it pertains to these processes, as well as discussing the therapeutic implications of these findings.


Assuntos
Cardiopatias/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração , Animais , Linhagem da Célula , Transdiferenciação Celular , Reprogramação Celular , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica , Cardiopatias/genética , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , MicroRNAs/genética , Miócitos Cardíacos/patologia , Fenótipo , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia
4.
Circ Res ; 116(3): 418-24, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25351576

RESUMO

RATIONALE: A major goal for the treatment of heart tissue damaged by cardiac injury is to develop strategies for restoring healthy heart muscle through the regeneration and repair of damaged myocardium. We recently demonstrated that administration of a specific combination of microRNAs (miR combo) into the infarcted myocardium leads to direct in vivo reprogramming of noncardiac myocytes to cardiac myocytes. However, the biological and functional consequences of such reprogramming are not yet known. OBJECTIVE: The aim of this study was to determine whether noncardiac myocytes directly reprogrammed using miRNAs in vivo develop into mature functional cardiac myocytes in situ, and whether reprogramming leads to improvement of cardiac function. METHODS AND RESULTS: We subjected fibroblast-specific protein 1-Cre mice/tandem dimer Tomato (tdTomato) mice to cardiac injury by permanent ligation of the left anterior descending coronary artery and injected lentiviruses encoding miR combo or a control nontargeting miRNA. miR combo significantly increased the number of reprogramming events in vivo. Five to 6 weeks after injury, morphological and physiological properties of tdTomato(-) and tdTomato(+) cardiac myocyte-like cells were analyzed ex vivo. tdTomato(+) cells expressed cardiac myocyte markers, sarcomeric organization, excitation-contraction coupling, and action potentials characteristic of mature ventricular cardiac myocytes (tdTomato(-) cells). Reprogramming was associated with improvement of cardiac function, as analyzed by serial echocardiography. There was a time delayed and progressive improvement in fractional shortening and other measures of ventricular function, indicating that miR combo promotes functional recovery of damaged myocardium. CONCLUSIONS: The findings from this study further validate the potential use of miRNA-mediated reprogramming as a therapeutic approach to promote cardiac regeneration after myocardial injury.


Assuntos
Reprogramação Celular , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Regeneração Tecidual Guiada , Masculino , Camundongos , MicroRNAs/genética , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Proteínas S100/metabolismo
5.
J Mol Cell Cardiol ; 85: 215-25, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26071893

RESUMO

Wnt signaling has recently emerged as an important regulator of cardiac progenitor cell proliferation and differentiation, but the exact mechanisms by which Wnt signaling modulates these effects are not known. Understanding these mechanisms is essential for advancing our knowledge of cardiac progenitor cell biology and applying this knowledge to enhance cardiac therapy. Here, we explored the effects of Sfrp2, a canonical Wnt inhibitor, in adult cardiac progenitor cell (CPC) differentiation and investigated the molecular mechanisms involved. Our data show that Sfrp2 treatment can promote differentiation of CPCs after ischemia-reperfusion injury. Treatment of CPCs with Sfrp2 inhibited CPC proliferation and primed them for cardiac differentiation. Sfrp2 binding to Wnt6 and inhibition of Wnt6 canonical pathway was essential for the inhibition of CPC proliferation. This inhibition of Wnt6 canonical signaling by Sfrp2 was important for activation of the non-canonical Wnt/Planar Cell Polarity (PCP) pathway through JNK, which in turn induced expression of cardiac transcription factors and CPC differentiation. Taken together, these results demonstrate a novel role of Sfrp2 and Wnt6 in regulating the dynamic process of CPC proliferation and differentiation, as well as providing new insights into the mechanisms of Wnt signaling in cardiac differentiation.


Assuntos
Diferenciação Celular , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Células-Tronco/fisiologia , Proteínas Wnt/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Expressão Gênica , Camundongos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Regulação para Cima , Via de Sinalização Wnt
6.
Circ Res ; 113(4): 372-80, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23784961

RESUMO

RATIONALE: The regenerative capacity of the heart is markedly diminished shortly after birth, coinciding with overall withdrawal of cardiomyocytes from cell cycle. Consequently, the adult mammalian heart has limited capacity to regenerate after injury. The discovery of factors that can induce cardiomyocyte proliferation is, therefore, of high interest and has been the focus of extensive investigation throughout the past years. OBJECTIVE: We have recently identified C3orf58 as a novel hypoxia and Akt induced stem cell factor (HASF) secreted from mesenchymal stem cells, which can promote cardiac repair through cytoprotective mechanisms. Here, we tested the hypothesis that HASF can also contribute to cardiac regeneration by stimulating cardiomyocyte division and proliferation. METHODS AND RESULTS: Neonatal ventricular cardiomyocytes were stimulated in culture for 7 days with purified recombinant HASF protein. Compared with control untreated cells, HASF-treated neonatal cardiomyocytes exhibited 60% increase in DNA synthesis as measured by bromodeoxyuridine incorporation. These results were confirmed by immunofluorescence confocal microscopy showing a 50% to 100% increase in the number of cardiomyocytes in the mitotic and cytokinesis phases. Importantly, in vivo cardiac overexpression of HASF in a transgenic mouse model resulted in enhanced level of DNA synthesis and cytokinesis in neonatal and adult cardiomyocytes. These proliferative effects were modulated by a phosphoinositide 3-kinase-protein kinase B-cycle-dependent kinase 7 pathway as revealed by the use of phosphoinositide 3-kinase -pathway-specific inhibitors and silencing of the Cdk7 gene. CONCLUSIONS: Our studies support the hypothesis that HASF induces cardiomyocyte proliferation via a phosphoinositide 3-kinase-protein kinase B-cycle-dependent kinase 7 pathway. The implications of this finding may be significant for cardiac regeneration biology and therapeutics.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/farmacologia , Ciclo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/fisiologia , Proteínas de Membrana/farmacologia , Miócitos Cardíacos/citologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Ciclo Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA/metabolismo , Coração/fisiologia , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Modelos Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Ratos , Proteínas Recombinantes/farmacologia , Regeneração , Transdução de Sinais/efeitos dos fármacos , Quinase Ativadora de Quinase Dependente de Ciclina
7.
J Mol Cell Cardiol ; 66: 157-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24269490

RESUMO

Despite advances in the treatment of acute tissue ischemia significant challenges remain in effective cytoprotection from ischemic cell death. It has been documented that injected stem cells, such as mesenchymal stem cells (MSCs), can confer protection to ischemic tissue through the release of paracrine factors. The study of these factors is essential for understanding tissue repair and the development of new therapeutic approaches for regenerative medicine. We have recently shown that a novel factor secreted by MSCs, which we called HASF (Hypoxia and Akt induced Stem cell Factor), promotes cardiomyocyte proliferation. In this study we show that HASF has a cytoprotective effect on ischemia induced cardiomyocyte death. We assessed whether HASF could potentially be used as a therapeutic agent to prevent the damage associated with myocardial infarction. In vitro treatment of cardiomyocytes with HASF protein resulted in decreased apoptosis; TUNEL positive nuclei were fewer in number, and caspase activation and mitochondrial pore opening were inhibited. Purified HASF protein was injected into the heart immediately following myocardial infarction. Heart function was found to be comparable to sham operated animals one month following injury and fibrosis was significantly reduced. In vivo and in vitro HASF activated protein kinase C ε (PKCε). Inhibition of PKCε blocked the HASF effect on apoptosis. Furthermore, the beneficial effects of HASF were lost in mice lacking PKCε. Collectively these results identify HASF as a protein of significant therapeutic potential, acting in part through PKCε.


Assuntos
Proteínas de Membrana/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo , Transdução de Sinais , Animais , Apoptose , Proliferação de Células/efeitos dos fármacos , Citoproteção , Regulação da Expressão Gênica , Humanos , Marcação In Situ das Extremidades Cortadas , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Comunicação Parácrina/genética , Proteína Quinase C-épsilon/genética
8.
Am J Physiol Renal Physiol ; 306(1): F12-23, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24197069

RESUMO

Renal damage resulting from acute and chronic kidney injury poses an important problem to public health. Currently, patients with end-stage renal disease rely solely on kidney transplantation or dialysis for survival. Emerging therapies aiming to prevent and reverse kidney damage are thus in urgent need. Although the kidney was initially thought to lack the capacity for self-repair, several studies have indicated that this might not be the case; progenitor and stem cells appear to play important roles in kidney repair under various pathological conditions. In this review, we summarize recent findings on the role of progenitor/stem cells on kidney repair as well as discuss their potential as a therapeutic approach for kidney diseases.


Assuntos
Insuficiência Renal/terapia , Transplante de Células-Tronco , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Regeneração/fisiologia
9.
Stem Cells ; 31(8): 1669-82, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23666637

RESUMO

Mesenchymal stem cells (MSCs) transplanted into injured myocardium promote repair through paracrine mechanisms. We have previously shown that MSCs over-expressing AKT1 (Akt-MSCs) exhibit enhanced properties for cardiac repair. In this study, we investigated the relevance of Abi3bp toward MSC biology. Abi3bp formed extracellular deposits with expression controlled by Akt1 and ubiquitin-mediated degradation. Abi3bp knockdown/knockout stabilized focal adhesions and promoted stress-fiber formation. Furthermore, MSCs from Abi3bp knockout mice displayed severe deficiencies in osteogenic and adipogenic differentiation. Knockout or stable knockdown of Abi3bp increased MSC and Akt-MSC proliferation, promoting S-phase entry via cyclin-d1, ERK1/2, and Src. Upon Abi3bp binding to integrin-ß1 Src associated with paxillin which inhibited proliferation. In vivo, Abi3bp knockout increased MSC number and proliferation in bone marrow, lung, and liver. In summary, we have identified a novel extracellular matrix protein necessary for the switch from proliferation to differentiation in MSCs.


Assuntos
Proteínas de Transporte/fisiologia , Comunicação Celular/fisiologia , Células-Tronco Mesenquimais/fisiologia , Animais , Comunicação Autócrina , Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Movimento Celular/fisiologia , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transfecção , Ubiquitina/metabolismo
10.
Circ Res ; 110(11): 1465-73, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539765

RESUMO

RATIONALE: Repopulation of the injured heart with new, functional cardiomyocytes remains a daunting challenge for cardiac regenerative medicine. An ideal therapeutic approach would involve an effective method at achieving direct conversion of injured areas to functional tissue in situ. OBJECTIVE: The aim of this study was to develop a strategy that identified and evaluated the potential of specific micro (mi)RNAs capable of inducing reprogramming of cardiac fibroblasts directly to cardiomyocytes in vitro and in vivo. METHODS AND RESULTS: Using a combinatorial strategy, we identified a combination of miRNAs 1, 133, 208, and 499 capable of inducing direct cellular reprogramming of fibroblasts to cardiomyocyte-like cells in vitro. Detailed studies of the reprogrammed cells demonstrated that a single transient transfection of the miRNAs can direct a switch in cell fate as documented by expression of mature cardiomyocyte markers, sarcomeric organization, and exhibition of spontaneous calcium flux characteristic of a cardiomyocyte-like phenotype. Interestingly, we also found that miRNA-mediated reprogramming was enhanced 10-fold on JAK inhibitor I treatment. Importantly, administration of miRNAs into ischemic mouse myocardium resulted in evidence of direct conversion of cardiac fibroblasts to cardiomyocytes in situ. Genetic tracing analysis using Fsp1Cre-traced fibroblasts from both cardiac and noncardiac cell sources strongly suggests that induced cells are most likely of fibroblastic origin. CONCLUSIONS: The findings from this study provide proof-of-concept that miRNAs have the capability of directly converting fibroblasts to a cardiomyocyte-like phenotype in vitro. Also of significance is that this is the first report of direct cardiac reprogramming in vivo. Our approach may have broad and important implications for therapeutic tissue regeneration in general.


Assuntos
Transdiferenciação Celular , Fibroblastos/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Transdiferenciação Celular/efeitos dos fármacos , Transdiferenciação Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Regulação da Expressão Gênica , Terapia Genética/métodos , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/administração & dosagem , Contração Miocárdica , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Inibidores de Proteínas Quinases/farmacologia , Recuperação de Função Fisiológica , Regeneração , Proteínas S100/genética , Proteínas S100/metabolismo , Transfecção , Proteína Vermelha Fluorescente
11.
J Am Soc Nephrol ; 24(8): 1263-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23744888

RESUMO

The renin-angiotensin-aldosterone system (RAAS) regulates BP and salt-volume homeostasis. Juxtaglomerular (JG) cells synthesize and release renin, which is the first and rate-limiting step in the RAAS. Intense pathologic stresses cause a dramatic increase in the number of renin-producing cells in the kidney, termed JG cell recruitment, but how this occurs is not fully understood. Here, we isolated renal CD44(+) mesenchymal stem cell (MSC)-like cells and found that they differentiated into JG-like renin-expressing cells both in vitro and in vivo. Sodium depletion and captopril led to activation and differentiation of these cells into renin-expressing cells in the adult kidney. In summary, CD44(+) MSC-like cells exist in the adult kidney and can differentiate into JG-like renin-producing cells under conditions that promote JG cell recruitment.


Assuntos
Células-Tronco Adultas/metabolismo , Captopril/farmacologia , Diferenciação Celular/fisiologia , Sistema Justaglomerular/citologia , Rim/citologia , Células-Tronco Mesenquimais/metabolismo , Sistema Renina-Angiotensina/fisiologia , Renina/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Sistema Justaglomerular/metabolismo , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Renina-Angiotensina/efeitos dos fármacos
12.
Respiration ; 85(2): 89-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23406722

RESUMO

Over the past few years, new insights have been added to the study of stem cells in the adult lung. The exploration of endogenous lung progenitors as well as the study of exogenously delivered stem cell populations holds promise for advancing our understanding of the biology of lung repair mechanisms. Moreover, it opens new possibilities for the use of stem cell therapy for the development of regenerative medicine approaches for the treatment of lung disease. Here, we discuss the main types of lung epithelial progenitor populations; the potential of endothelial progenitors, mesenchymal stem cells and embryonic stem cells for lung therapy, as well as summarize the cellular mechanisms involved.


Assuntos
Células-Tronco Adultas/fisiologia , Pneumopatias/terapia , Pulmão/citologia , Adulto , Células-Tronco Embrionárias/fisiologia , Endotélio Vascular/citologia , Humanos , Células-Tronco Mesenquimais/fisiologia , Regeneração/fisiologia , Medicina Regenerativa , Transplante de Células-Tronco , Engenharia Tecidual
13.
Proc Natl Acad Sci U S A ; 107(49): 21110-5, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21078975

RESUMO

Secreted frizzled related protein 2 (Sfrp2) is known as an inhibitor for the Wnt signaling. In recent studies, Sfrp2 has been reported to inhibit the activity of Xenopus homolog of mammalian Tolloid-like 1 metalloproteinase. Bone morphogenic protein 1 (Bmp1)/Tolloid-like metalloproteinase plays a key role in the regulation of collagen biosynthesis and maturation after tissue injury. Here, we showed both endogenous Sfrp2 and Bmp1 protein expressions were up-regulated in rat heart after myocardial infarction (MI). We hypothesize that Sfrp2 could inhibit mammalian Bmp1 activity and, hence, the exogenous administration of Sfrp2 after MI would inhibit the deposition of mature collagen and improve heart function. Using recombinant proteins, we demonstrated that Sfrp2, but not Sfrp1 or Sfrp3, inhibited Bmp1 activity in vitro as measured by a fluorogenic peptide based procollagen C-proteinase activity assay. We also demonstrated that Sfrp2 at high concentration inhibited human and rat type I procollagen processing by Bmp1 in vitro. We further showed that exogenously added Sfrp2 inhibited type I procollagen maturation in primary cardiac fibroblasts. Two days after direct injection into the rat infarcted myocardium, Sfrp2 inhibited MI-induced type I collagen deposition. As early as 2 wk after injection, Sfrp2 significantly reduced left ventricular (LV) fibrosis as shown by trichrome staining. Four weeks after injection, Sfrp2 prevented the anterior wall thinning and significantly improved cardiac function as revealed by histological analysis and echocardiographic measurement. Our study demonstrates Sfrp2 at therapeutic doses can inhibit fibrosis and improve LV function at a later stage after MI.


Assuntos
Fibrose/tratamento farmacológico , Receptores Frizzled/análise , Receptores Frizzled/uso terapêutico , Proteínas de Membrana/uso terapêutico , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Proteína Morfogenética Óssea 1/análise , Proteína Morfogenética Óssea 1/antagonistas & inibidores , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Receptores Frizzled/administração & dosagem , Humanos , Masculino , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/análise , Infarto do Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Disfunção Ventricular Esquerda/tratamento farmacológico
14.
Front Cardiovasc Med ; 10: 1096884, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37283579

RESUMO

Scalable and high-throughput electrophysiological measurement systems are necessary to accelerate the elucidation of cardiac diseases in drug development. Optical mapping is the primary method of simultaneously measuring several key electrophysiological parameters, such as action potentials, intracellular free calcium and conduction velocity, at high spatiotemporal resolution. This tool has been applied to isolated whole-hearts, whole-hearts in-vivo, tissue-slices and cardiac monolayers/tissue-constructs. Although optical mapping of all of these substrates have contributed to our understanding of ion-channels and fibrillation dynamics, cardiac monolayers/tissue-constructs are scalable macroscopic substrates that are particularly amenable to high-throughput interrogation. Here, we describe and validate a scalable and fully-automated monolayer optical mapping robot that requires no human intervention and with reasonable costs. As a proof-of-principle demonstration, we performed parallelized macroscopic optical mapping of calcium dynamics in the well-established neonatal-rat-ventricular-myocyte monolayer plated on standard 35 mm dishes. Given the advancements in regenerative and personalized medicine, we also performed parallelized macroscopic optical mapping of voltage dynamics in human pluripotent stem cell-derived cardiomyocyte monolayers using a genetically encoded voltage indictor and a commonly-used voltage sensitive dye to demonstrate the versatility of our system.

15.
Circ Res ; 106(11): 1753-62, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20378860

RESUMO

RATIONALE: Although mesenchymal stem cell (MSC) transplantation has been shown to promote cardiac repair in acute myocardial injury in vivo, its overall restorative capacity appears to be restricted mainly because of poor cell viability and low engraftment in the ischemic myocardium. Specific chemokines are upregulated in the infarcted myocardium. However the expression levels of the corresponding chemokine receptors (eg, CCR1, CXCR2) in MSCs are very low. We hypothesized that this discordance may account for the poor MSC engraftment and survival. OBJECTIVE: To determine whether overexpression of CCR1 or CXCR2 chemokine receptors in MSCs augments their cell survival, migration and engraftment after injection in the infarcted myocardium. METHODS AND RESULTS: Overexpression of CCR1, but not CXCR2, dramatically increased chemokine-induced murine MSC migration and protected MSC from apoptosis in vitro. Moreover, when MSCs were injected intramyocardially one hour after coronary artery ligation, CCR1-MSCs accumulated in the infarcted myocardium at significantly higher levels than control-MSCs or CXCR2-MSCs 3 days postmyocardial infarction (MI). CCR1-MSC-injected hearts exhibited a significant reduction in infarct size, reduced cardiomyocytes apoptosis and increased capillary density in injured myocardium 3 days after MI. Furthermore, intramyocardial injection of CCR1-MSCs prevented cardiac remodeling and restored cardiac function 4 weeks after MI. CONCLUSIONS: Our results demonstrate the in vitro and in vivo salutary effects of genetic modification of stem cells. Specifically, overexpression of chemokine receptor enhances the migration, survival and engraftment of MSCs, and may provide a new therapeutic strategy for the injured myocardium.


Assuntos
Capilares/metabolismo , Movimento Celular , Células Endoteliais/metabolismo , Terapia Genética/métodos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Neovascularização Fisiológica , Receptores CCR1/biossíntese , Animais , Apoptose , Capilares/patologia , Capilares/fisiopatologia , Sobrevivência Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Fibrose , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Interferência de RNA , Receptores CCR1/genética , Receptores de Interleucina-8B/biossíntese , Receptores de Interleucina-8B/genética , Recuperação de Função Fisiológica , Regeneração , Fatores de Tempo , Transdução Genética , Função Ventricular Esquerda , Remodelação Ventricular
16.
J Mol Cell Cardiol ; 50(2): 280-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20727900

RESUMO

Stem cells play an important role in restoring cardiac function in the damaged heart. In order to mediate repair, stem cells need to replace injured tissue by differentiating into specialized cardiac cell lineages and/or manipulating the cell and molecular mechanisms governing repair. Despite early reports describing engraftment and successful regeneration of cardiac tissue in animal models of heart failure, these events appear to be infrequent and yield too few new cardiomyocytes to account for the degree of improved cardiac function observed. Instead, mounting evidence suggests that stem cell mediated repair takes place via the release of paracrine factors into the surrounding tissue that subsequently direct a number of restorative processes including myocardial protection, neovascularization, cardiac remodeling, and differentiation. The potential for diverse stem cell populations to moderate many of the same processes as well as key paracrine factors and molecular pathways involved in stem cell-mediated cardiac repair will be discussed in this review. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Assuntos
Coração/fisiologia , Comunicação Parácrina , Regeneração , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Coração/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico
17.
Physiol Genomics ; 27(1): 86-94, 2006 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-16835353

RESUMO

The pathogenesis of cardiac left ventricular hypertrophy and failure is poorly defined due to the complexity of the disease phenotype. To gain a better understanding of the relationship between gene expression and left ventricular hypertrophy, we employed a quantitative approach to identify genes with expression patterns that correlate in a numerically continuous manner with parameters of cardiac structure and function in a mouse model of left ventricular hypertrophy due to transverse aortic constriction. Several genes showed expression patterns that were significantly correlated (Pearson's correlation coefficient) with measurements of left ventricular weight, left ventricular wall thickness, and diastolic dimension. We validated our findings in two independent data sets and in a small subset of genes by real-time RT-PCR. Of genes with significant correlations to numerically continuous measurements of hypertrophy, we found enrichment for genes encoding extracellular matrix, growth-related and secreted proteins in the directly correlated subset, and for genes encoding mitochondria and metabolic/fatty acid oxidation proteins in the inversely correlated subset. The results of this filtering strategy suggest that this subset of transcripts with quantitative relationships between gene expression and left ventricular hypertrophy represents potentially important pathways that contribute to the progression to heart failure and are thus candidates for follow-up and functional analysis.


Assuntos
Expressão Gênica , Hipertrofia Ventricular Esquerda/genética , Animais , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica , Genômica , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/metabolismo , Masculino , Metanálise como Assunto , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Ultrassonografia
18.
Methods Mol Biol ; 1416: 495-507, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27236692

RESUMO

Mesenchymal stem cells (MSC) from bone marrow or adult tissues are widely studied to evaluate their potential for tissue repair. Differences in tissue of origin, donor variation, or in vitro handling exist and it is still unclear how they affect cell function and regenerative potential. Large-scale gene expression analysis of these cells not only allows researchers to compare and contrast the differences between each MSC subset but also allows for the development of better analytical tools for their characterization and utilization. Here, we describe a protocol for transcriptomics analysis of MSC-like cells derived from adult kidneys.


Assuntos
Células-Tronco Adultas/metabolismo , Perfilação da Expressão Gênica/métodos , Rim/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Adultas/citologia , Animais , Separação Celular , Células Cultivadas , Criopreservação , Rim/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos
19.
Sci Rep ; 6: 38815, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27941896

RESUMO

We have recently shown that a combination of microRNAs, miR combo, can directly reprogram cardiac fibroblasts into functional cardiomyocytes in vitro and in vivo. Reprogramming of cardiac fibroblasts by miR combo in vivo is associated with improved cardiac function following myocardial infarction. However, the efficiency of direct reprogramming in vitro is relatively modest and new strategies beyond the traditional two-dimensional (2D) culture should be identified to improve reprogramming process. Here, we report that a tissue-engineered three-dimensional (3D) hydrogel environment enhanced miR combo reprogramming of neonatal cardiac and tail-tip fibroblasts. This was associated with significantly increased MMPs expression in 3D vs. 2D cultured cells, while pharmacological inhibition of MMPs blocked the effect of the 3D culture on enhanced miR combo mediated reprogramming. We conclude that 3D tissue-engineered environment can enhance the direct reprogramming of fibroblasts to cardiomyocytes via a MMP-dependent mechanism.


Assuntos
Microambiente Celular/fisiologia , Reprogramação Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , MicroRNAs/farmacologia , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Animais , Animais Recém-Nascidos , Células Cultivadas , Reprogramação Celular/fisiologia , Fibroblastos/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter , Hidrogéis , Camundongos , MicroRNAs/genética , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transfecção
20.
J Biophotonics ; 9(4): 406-13, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27095616

RESUMO

High-resolution tracking of stem cells remains a challenging task. An ultra-bright contrast agent with extended intracellular retention is suitable for in vivo high-resolution tracking of stem cells following the implantation. Here, a plasmonic-active nanoplatform was developed for tracking mesenchymal stromal cells (MSCs) in mice. The nanoplatform consisted of TAT peptide-functionalized gold nanostars (TAT-GNS) that emit ultra-bright two-photon photoluminescence capable of tracking MSCs under high-resolution optical imaging. In vitro experiment showed TAT-GNS-labeled MSCs retained a similar differentiability to that of non-labeled MSCs controls. Due to their star shape, TAT-GNS exhibited greater intracellular retention than that of commercial Q-Tracker. In vivo imaging of TAT-GNS-labeled MSCs five days following intra-arterial injections in mice kidneys showed possible MSCs implantation in juxta-glomerular (JG) regions, but non-specifically in glomeruli and afferent arterioles as well. With future design to optimize GNS labeling specificity and clearance, plasmonic-active nanoplatforms may be a useful intracellular tracking tool for stem cell research. An ultra-bright intracellular contrast agent is developed using TAT peptide-functionalized gold nanostars (TAT-GNS). It poses minimal influence on the stem cell differentiability. It exhibits stronger two-photon photoluminescence and superior labeling efficiency than commercial Q-Tracker. Following renal implantation, some TAT-GNS-labeled MSCs permeate blood vessels and migrate to the juxta-glomerular region.


Assuntos
Rastreamento de Células/métodos , Produtos do Gene tat/química , Células-Tronco Mesenquimais/citologia , Nanotecnologia/métodos , Animais , Diferenciação Celular/efeitos dos fármacos , Ouro/química , Rim/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanoestruturas/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa