Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Dev Cell ; 56(19): 2722-2740.e6, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34610329

RESUMO

Spiny mice (Acomys cahirinus) are terrestrial mammals that evolved unique scar-free regenerative wound-healing properties. Myofibroblasts (MFs) are the major scar-forming cell type in skin. We found that following traumatic injury to ear pinnae, MFs appeared rapidly in both Acomys and mouse yet persisted only in mouse. The timing of MF loss in Acomys correlated with wound closure, blastema differentiation, and nuclear localization of the Hippo pathway target protein Yap. Experiments in vitro revealed an accelerated PP2A-dependent dephosphorylation activity that maintained nuclear Yap in Acomys dermal fibroblasts (DFs) and was not detected in mouse or human DFs. Treatment of Acomys in vivo with the nuclear Yap-TEAD inhibitor verteporfin prolonged MF persistence and converted tissue regeneration to fibrosis. Forced Yap activity prevented and rescued TGF-ß1-induced human MF formation in vitro. These results suggest that Acomys evolved modifications of Yap activity and MF fate important for scar-free regenerative wound healing in vivo.


Assuntos
Via de Sinalização Hippo/fisiologia , Cicatrização/fisiologia , Proteínas de Sinalização YAP/metabolismo , Animais , Cicatriz/metabolismo , Cicatriz/patologia , Orelha/patologia , Camundongos , Murinae/fisiologia , Miofibroblastos/metabolismo , Pele/metabolismo
3.
Circ Res ; 120(2): 296-311, 2017 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-27834190

RESUMO

RATIONALE: The vascular adventitia is a complex layer of the vessel wall consisting of vasa vasorum microvessels, nerves, fibroblasts, immune cells, and resident progenitor cells. Adventitial progenitors express the stem cell markers, Sca1 and CD34 (adventitial sca1-positive progenitor cells [AdvSca1]), have the potential to differentiate in vitro into multiple lineages, and potentially contribute to intimal lesions in vivo. OBJECTIVE: Although emerging data support the existence of AdvSca1 cells, the goal of this study was to determine their origin, degree of multipotency and heterogeneity, and contribution to vessel remodeling. METHODS AND RESULTS: Using 2 in vivo fate-mapping approaches combined with a smooth muscle cell (SMC) epigenetic lineage mark, we report that a subpopulation of AdvSca1 cells is generated in situ from differentiated SMCs. Our data establish that the vascular adventitia contains phenotypically distinct subpopulations of progenitor cells expressing SMC, myeloid, and hematopoietic progenitor-like properties and that differentiated SMCs are a source to varying degrees of each subpopulation. SMC-derived AdvSca1 cells exhibit a multipotent phenotype capable of differentiating in vivo into mature SMCs, resident macrophages, and endothelial-like cells. After vascular injury, SMC-derived AdvSca1 cells expand in number and are major contributors to adventitial remodeling. Induction of the transcription factor Klf4 in differentiated SMCs is essential for SMC reprogramming in vivo, whereas in vitro approaches demonstrate that Klf4 is essential for the maintenance of the AdvSca1 progenitor phenotype. CONCLUSIONS: We propose that generation of resident vascular progenitor cells from differentiated SMCs is a normal physiological process that contributes to the vascular stem cell pool and plays important roles in arterial homeostasis and disease.


Assuntos
Túnica Adventícia/citologia , Túnica Adventícia/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Feminino , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos de Músculo Liso/fisiologia , Gravidez
4.
J Biol Chem ; 290(16): 10353-67, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25733666

RESUMO

This study identifies signaling pathways that play key roles in the formation and maintenance of epicardial cells, a source of progenitors for coronary smooth muscle cells (SMCs). After epithelial to mesenchymal transition (EMT), mesenchymal cells invade the myocardium to form coronary SMCs. RhoA/Rho kinase activity is required for EMT and for differentiation into coronary SMCs, whereas cAMP activity is known to inhibit EMT in epithelial cells by an unknown mechanism. We use outgrowth of epicardial cells from E9.5 isolated mouse proepicardium (PE) explants, wild type and Epac1 null E12.5 mouse heart explants, adult rat epicardial cells, and immortalized mouse embryonic epicardial cells as model systems to identify signaling pathways that regulate RhoA activity to maintain the epicardial progenitor state. We demonstrate that RhoA activity is suppressed in the epicardial progenitor state, that the cAMP-dependent Rap1 GTP exchange factor (GEF), Epac, known to down-regulate RhoA activity through activation of Rap1 GTPase activity increased, that Rap1 activity increased, and that expression of the RhoA antagonistic Rnd proteins known to activate p190RhoGAP increased and associated with p190RhoGAP. Finally, EMT is associated with increased p63RhoGEF and RhoGEF-H1 protein expression, increased GEF-H1 activity, with a trend in increased p63RhoGEF activity. EMT is suppressed by partial silencing of p63RhoGEF and GEF-H1. In conclusion, we have identified new signaling molecules that act together to control RhoA activity and play critical roles in the maintenance of coronary smooth muscle progenitor cells in the embryonic epicardium. We suggest that their eventual manipulation could promote revascularization after myocardial injury.


Assuntos
Miócitos de Músculo Liso/metabolismo , Pericárdio/metabolismo , Células-Tronco/metabolismo , Proteínas rho de Ligação ao GTP/genética , Animais , Diferenciação Celular , Embrião de Mamíferos , Transição Epitelial-Mesenquimal/genética , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Miócitos de Músculo Liso/citologia , Pericárdio/citologia , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Técnicas de Cultura de Tecidos , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
5.
J Dev Biol ; 3(4): 93-111, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26770887

RESUMO

Proper control of the temporal onset of cellular differentiation is critical for regulating cell lineage decisions and morphogenesis during development. Pbx homeodomain transcription factors have emerged as important regulators of cellular differentiation. We previously showed, by using antisense morpholino knockdown, that Pbx factors are needed for the timely activation of myocardial differentiation in zebrafish. In order to gain further insight into the roles of Pbx factors in heart development, we show here that zebrafish pbx4 mutant embryos exhibit delayed onset of myocardial differentiation, such as delayed activation of tnnt2a expression in early cardiomyocytes in the anterior lateral plate mesoderm. We also observe delayed myocardial morphogenesis and dysmorphic patterning of the ventricle and atrium, consistent with our previous Pbx knock-down studies. In addition, we find that pbx4 mutant larvae have aberrant outflow tracts and defective expression of the proepicardial marker tbx18. Finally, we present evidence for Pbx expression in cardiomyocyte precursors as well as heterogeneous Pbx expression among the pan-cytokeratin-expressing proepicardial cells near the developing ventricle. In summary, our data show that Pbx4 is required for the proper temporal activation of myocardial differentiation and establish a basis for studying additional roles of Pbx factors in heart development.

6.
Dev Biol ; 383(2): 307-20, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24016759

RESUMO

The epicardium and coronary vessels originate from progenitor cells in the proepicardium. Here we show that Tbx18, a T-box family member highly expressed in the proepicardium, controls critical early steps in coronary development. In Tbx18(-/-) mouse embryos, both the epicardium and coronary vessels exhibit structural and functional defects. At E12.5, the Tbx18-deficient epicardium contains protrusions and cyst-like structures overlying a disorganized coronary vascular plexus that contains ectopic structures resembling blood islands. At E13.5, the left and right coronary stems form correctly in mutant hearts. However, analysis of PECAM-1 whole mount immunostaining, distribution of SM22α(lacZ/+) activity, and analysis of coronary vascular casts suggest that defective vascular plexus remodeling produces a compromised arterial network at birth consisting of fewer distributing conduit arteries with smaller lumens and a reduced capacity to conduct blood flow. Gene expression profiles of Tbx18(-/-) hearts at E12.5 reveal altered expression of 79 genes that are associated with development of the vascular system including sonic hedgehog signaling components patched and smoothened, VEGF-A, angiopoietin-1, endoglin, and Wnt factors compared to wild type hearts. Thus, formation of coronary vasculature is responsive to Tbx18-dependent gene targets in the epicardium, and a poorly structured network of coronary conduit vessels is formed in Tbx18 null hearts due to defects in epicardial cell signaling and fate during heart development. Lastly, we demonstrate that Tbx18 possesses a SRF/CArG box dependent repressor activity capable of inhibiting progenitor cell differentiation into smooth muscle cells, suggesting a potential function of Tbx18 in maintaining the progenitor status of epicardial-derived cells.


Assuntos
Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Pericárdio/embriologia , Pericárdio/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Circulação Coronária , Vasos Coronários/patologia , Vasos Coronários/ultraestrutura , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Miócitos de Músculo Liso/metabolismo , Pericárdio/patologia , Pericárdio/ultraestrutura , Proteínas Repressoras/metabolismo , Fator de Resposta Sérica/química , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Transcrição Gênica , beta-Galactosidase/metabolismo
7.
PLoS One ; 8(4): e61421, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23637832

RESUMO

Hedgehog (Hh) signaling plays fundamental roles in morphogenesis, tissue repair, and human disease. Initiation of Hh signaling is controlled by the interaction of two multipass membrane proteins, patched (Ptc) and smoothened (Smo). Recent studies identify Smo as a G-protein coupled receptor (GPCR)-like protein that signals through large G-protein complexes which contain the Gαi subunit. We hypothesize Regulator of G-Protein Signaling (RGS) proteins, and specifically RGS5, are endogenous repressors of Hh signaling via their ability to act as GTPase activating proteins (GAPs) for GTP-bound Gαi, downstream of Smo. In support of this hypothesis, we demonstrate that RGS5 over-expression inhibits sonic hedgehog (Shh)-mediated signaling and osteogenesis in C3H10T1/2 cells. Conversely, signaling is potentiated by siRNA-mediated knock-down of RGS5 expression, but not RGS4 expression. Furthermore, using immuohistochemical analysis and co-immunoprecipitation (Co-IP), we demonstrate that RGS5 is present with Smo in primary cilia. This organelle is required for canonical Hh signaling in mammalian cells, and RGS5 is found in a physical complex with Smo in these cells. We therefore conclude that RGS5 is an endogenous regulator of Hh-mediated signaling and that RGS proteins are potential targets for novel therapeutics in Hh-mediated diseases.


Assuntos
Proteínas RGS/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Cílios/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Técnicas de Silenciamento de Genes , Proteínas Hedgehog/efeitos dos fármacos , Camundongos , Proteínas RGS/biossíntese , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Receptor Smoothened
9.
Cells Tissues Organs ; 195(1-2): 73-81, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22005572

RESUMO

Recent observations suggest that the adventitial layer of blood vessels exhibits properties resembling a stem/progenitor cell niche. Progenitor cells have been isolated from the adventitia of both murine and human blood vessels with the potential to form endothelial cells, mural cells, osteogenic cells, and adipocytes. These progenitors appear to cluster at or near the border zone between the outer media and inner adventitia. In the mouse, this border zone region corresponds to a localized site of sonic hedgehog signaling in the artery wall. This brief review will discuss the emerging evidence that the tunica adventitia may provide a niche-like signaling environment for resident progenitor cells and will address the role of the adventitia in growth, remodeling, and repair of the artery wall.


Assuntos
Células do Tecido Conjuntivo/citologia , Tecido Conjuntivo/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Animais , Artérias/citologia , Artérias/fisiologia , Artérias/fisiopatologia , Tecido Conjuntivo/fisiopatologia , Humanos
10.
Arterioscler Thromb Vasc Biol ; 31(7): 1530-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21677296

RESUMO

Conventional views of the tunica adventitia as a poorly organized layer of vessel wall composed of fibroblasts, connective tissue, and perivascular nerves are undergoing revision. Recent studies suggest that the adventitia has properties of a stem/progenitor cell niche in the artery wall that may be poised to respond to arterial injury. It is also a major site of immune surveillance and inflammatory cell trafficking and harbors a dynamic microvasculature, the vasa vasorum, that maintains the medial layer and provides an important gateway for macrophage and leukocyte migration into the intima. In addition, the adventitia is in contact with tissue that surrounds the vessel and may actively participate in exchange of signals and cells between the vessel wall and the tissue in which it resides. This brief review highlights recent advances in our understanding of the adventitia and its resident progenitor cells and discusses progress toward an integrated view of adventitial function in vascular development, repair, and disease.


Assuntos
Tecido Conjuntivo/patologia , Células-Tronco/patologia , Doenças Vasculares/patologia , Animais , Comunicação Celular , Tecido Conjuntivo/imunologia , Tecido Conjuntivo/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Fenótipo , Transdução de Sinais , Nicho de Células-Tronco , Células-Tronco/imunologia , Células-Tronco/metabolismo , Doenças Vasculares/imunologia , Doenças Vasculares/metabolismo
12.
Circ Res ; 108(3): 365-77, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21293008

RESUMO

Molecular pathways that control the specification, migration, and number of available smooth muscle progenitor cells play key roles in determining blood vessel size and structure, capacity for tissue repair, and progression of age-related disorders. Defects in these pathways produce malformations of developing blood vessels, depletion of smooth muscle progenitor cell pools for vessel wall maintenance and repair, and aberrant activation of alternative differentiation pathways in vascular disease. A better understanding of the molecular mechanisms that uniquely specify and maintain vascular smooth muscle cell precursors is essential if we are to use advances in stem and progenitor cell biology and somatic cell reprogramming for applications directed to the vessel wall.


Assuntos
Vasos Sanguíneos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Músculo Liso Vascular/fisiologia , Animais , Vasos Sanguíneos/citologia , Diferenciação Celular/fisiologia , Epigenômica , Humanos , Células-Tronco Mesenquimais/citologia , Músculo Liso Vascular/citologia , Regeneração/fisiologia
13.
Atherosclerosis ; 215(1): 34-42, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20810113

RESUMO

OBJECTIVE: Nicotinic acetylcholine receptor α1 (nAChRα1) was recently identified as a functional cell receptor for urokinase, a potent atherogenic molecule. Here, we test the hypothesis that nAChRα1 plays a role in the pathogenesis of atherosclerosis. METHODS: Apolipoprotein E-deficient mice were initially fed a Western diet for 8 wks. Plasmid DNA encoding scramble RNA (pscr) or siRNA (psir2) for nAChRα1 was injected into the mice (n=16) using an aortic hydrodynamic gene transfer protocol. Four mice from each group were sacrificed 7 days after the DNA injection to confirm the nAChRα1 gene silencing. The remaining mice continued on a Western diet for an additional 16 wks. RESULTS: The nAChRα1 was up-regulated in aortic atherosclerotic lesions. A 78% knockdown of the nAChRα1 gene resulted in remarkably less severe aortic plaque growth and neovascularization at 16 wks (both P<0.05). In addition, significantly fewer macrophages (60% less) and myofibroblasts (80% less) presented in the atherosclerotic lesion of the psir2-treated mice. The protective mechanisms of the nAChRα1 knockdown may involve up-regulating interferon-γ/Y box protein-1 activity and down-regulating transforming growth factor-ß expression. CONCLUSIONS: The nAChRα1 gene plays a significant role at the artery wall, and reducing its expression decreases aortic plaque development.


Assuntos
Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Receptores Nicotínicos/genética , Actinas/biossíntese , Animais , Aorta/metabolismo , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Feminino , Inativação Gênica , Interferon gama/biossíntese , Macrófagos/metabolismo , Camundongos , Miofibroblastos/metabolismo , Fatores de Transcrição/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Regulação para Cima
14.
Arterioscler Thromb Vasc Biol ; 30(12): 2575-86, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20884876

RESUMO

OBJECTIVE: Congenital heart defects represent the most common human birth defects. Even though the genetic cause of these syndromes has been linked to candidate genes, the underlying molecular mechanisms are still largely unknown. Disturbance of neural crest cell (NCC) migration into the derivatives of the pharyngeal arches and pouches can account for many of the developmental defects. The goal of this study was to investigate the function of microRNA (miRNA) in NCCs and the cardiovascular system. METHODS AND RESULTS: We deleted Dicer from the NCC lineage and showed that Dicer conditional mutants exhibit severe defects in multiple craniofacial and cardiovascular structures, many of which are observed in human neuro-craniofacial-cardiac syndrome patients. We found that cranial NCCs require Dicer for their survival and that deletion of Dicer led to massive cell death and complete loss of NCC-derived craniofacial structures. In contrast, Dicer and miRNAs were not essential for the survival of cardiac NCCs. However, the migration and patterning of these cells were impaired in Dicer knockout mice, resulting in a spectrum of cardiovascular abnormalities, including type B interrupted aortic arch, double-outlet right ventricle, and ventricular septal defect. We showed that Dicer loss of function was, at least in part, mediated by miRNA-21 (miR-21) and miRNA-181a (miR-181a), which in turn repressed the protein level of Sprouty 2, an inhibitor of Erk1/2 signaling. CONCLUSIONS: Our results uncovered a central role for Dicer and miRNAs in NCC survival, migration, and patterning in craniofacial and cardiovascular development which, when mutated, lead to congenital neuro-craniofacial-cardiac defects.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Craniofaciais/genética , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , MicroRNAs/metabolismo , Crista Neural/metabolismo , Ribonuclease III/genética , Anormalidades Múltiplas/embriologia , Anormalidades Múltiplas/patologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Morte Celular , Diferenciação Celular , Movimento Celular , Sobrevivência Celular , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/patologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genótipo , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Crista Neural/patologia , Fenótipo , Proteínas Serina-Treonina Quinases , Ribonuclease III/deficiência , Índice de Gravidade de Doença , Síndrome
16.
Methods Enzymol ; 445: 209-28, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19022061

RESUMO

This chapter summarizes experimental techniques used to study coronary vessel development from its origins in the proepicardium (PE) to the final assembled network of arteries, veins, and capillaries present in the mature heart. Methods are described for microdissection and culture of the PE and embryonic epicardial cells, isolation of total RNA from single PE primordia and analysis by RT-PCR, imaging of the epicardium and coronary vessels by whole-mount confocal microscopy and by scanning electron microscopy, and the preparation of coronary vascular corrosion casts to visualize the entire coronary artery network structure. These techniques form the basic tools to study the cellular and molecular pathways that guide development and remodeling of coronary vessels.


Assuntos
Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Animais , Aves/embriologia , Vasos Coronários/ultraestrutura , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Embrião não Mamífero/ultraestrutura , Microscopia Confocal , Microscopia Eletrônica de Varredura , Pericárdio/embriologia , Pericárdio/metabolismo , Pericárdio/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Proc Natl Acad Sci U S A ; 105(27): 9349-54, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18591670

RESUMO

We characterize a sonic hedgehog (Shh) signaling domain restricted to the adventitial layer of artery wall that supports resident Sca1-positive vascular progenitor cells (AdvSca1). Using patched-1 (Ptc1(lacZ)) and patched-2 (Ptc2(lacZ)) reporter mice, adventitial Shh signaling activity was first detected at embryonic day (E) 15.5, reached the highest levels between postnatal day 1 (P1) and P10, was diminished in adult vessels, and colocalized with a circumferential ring of Shh protein deposited between the media and adventitia. In Shh(-/-) mice, AdvSca1 cells normally found at the aortic root were either absent or greatly diminished in number. Using a Wnt1-cre lineage marker that identifies cells of neural crest origin, we found that neither the adventitia nor AdvSca1 cells were labeled in arteries composed of neural crest-derived smooth muscle cells (SMCs). Although AdvSca1 cells do not express SMC marker proteins in vivo, they do express transcription factors thought to be required for SMC differentiation, including serum response factor (SRF) and myocardin family members, and readily differentiate to SMC-like cells in vitro. However, AdvSca1 cells also express potent repressors of SRF-dependent transcription, including Klf4, Msx1, and FoxO4, which may be critical for maintenance of the SMC progenitor phenotype of AdvSca1 cells in vivo. We conclude that a restricted domain of Shh signaling is localized to the arterial adventitia and may play important roles in maintenance of resident vascular SMC progenitor cells in the artery wall.


Assuntos
Artérias/metabolismo , Tecido Conjuntivo/metabolismo , Proteínas Hedgehog/metabolismo , Miócitos de Músculo Liso/citologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Aorta/citologia , Aorta/embriologia , Artérias/citologia , Artérias/embriologia , Ataxina-1 , Ataxinas , Separação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Proteínas Hedgehog/deficiência , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Células-Tronco/metabolismo
18.
Dev Cell ; 4(1): 107-18, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12530967

RESUMO

Cysteine-rich LIM-only proteins, CRP1 and CRP2, expressed during cardiovascular development act as bridging molecules that associate with serum response factor and GATA proteins. SRF-CRP-GATA complexes strongly activated smooth muscle gene targets. CRP2 was found in the nucleus during early stages of coronary smooth muscle differentiation from proepicardial cells. A dominant-negative CRP2 mutant blocked proepicardial cells from differentiating into smooth muscle cells. Together with SRF and GATA proteins, CRP1 and CRP2 converted pluripotent 10T1/2 fibroblasts into smooth muscle cells, while muscle LIM protein CRP3 inhibited the conversion. Thus, LIM-only proteins of the CRP family play important roles in organizing multiprotein complexes, both in the cytoplasm, where they participate in cytoskeletal remodeling, and in the nucleus, where they strongly facilitate smooth muscle differentiation.


Assuntos
Proteínas Aviárias , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína beta Intensificadora de Ligação a CCAAT/química , Proteína beta Intensificadora de Ligação a CCAAT/genética , Sistema Cardiovascular/citologia , Sistema Cardiovascular/embriologia , Proteínas de Transporte/química , Proteínas de Transporte/genética , Embrião de Galinha , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Fatores de Ligação de DNA Eritroide Específicos , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Proteínas com Domínio LIM , Substâncias Macromoleculares , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Resposta Sérica/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA