Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Cell Sci ; 130(14): 2382-2393, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28584191

RESUMO

The tyrosine kinase Src acts as a key regulator of cell motility by phosphorylating multiple protein substrates that control cytoskeletal and adhesion dynamics. In an earlier phosphotyrosine proteomics study, we identified a novel Rho-GTPase activating protein, now known as ARHGAP42, as a likely biologically relevant Src substrate. ARHGAP42 is a member of a family of RhoGAPs distinguished by tandem BAR-PH domains lying N-terminal to the GAP domain. Like other family members, ARHGAP42 acts preferentially as a GAP for RhoA. We show that Src principally phosphorylates ARHGAP42 on tyrosine 376 (Tyr-376) in the short linker between the BAR-PH and GAP domains. The expression of ARHGAP42 variants in mammalian cells was used to elucidate its regulation. We found that the BAR domain is inhibitory toward the GAP activity of ARHGAP42, such that BAR domain deletion resulted in decreased active GTP-bound RhoA and increased cell motility. With the BAR domain intact, ARHGAP42 GAP activity could be activated by phosphorylation of Tyr-376 to promote motile cell behavior. Thus, phosphorylation of ARHGAP42 Tyr-376 is revealed as a novel regulatory event by which Src can affect actin dynamics through RhoA inhibition.


Assuntos
Movimento Celular/fisiologia , Adesões Focais/metabolismo , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Tirosina/metabolismo , Quinases da Família src/metabolismo , Animais , Humanos , Camundongos , Fosforilação , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 315(6): H1614-H1626, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30359089

RESUMO

Cadherin-11 (CDH11) is upregulated in a variety of fibrotic diseases, including arthritis and calcific aortic valve disease. Our recent work has identified CDH11 as a potential therapeutic target and shown that treatment with a CDH11 functional blocking antibody can prevent hallmarks of calcific aortic valve disease in mice. The present study investigated the role of CDH11 in regulating the mechanobiological behavior of valvular interstitial cells believed to cause calcification. Aortic valve interstitial cells were harvested from Cdh11+/+, Cdh11+/-, and Cdh11-/- immortomice. Cells were subjected to inflammatory cytokines transforming growth factor (TGF)-ß1 and IL-6 to characterize the molecular mechanisms by which CDH11 regulates their mechanobiological changes. Histology was performed on aortic valves from Cdh11+/+, Cdh11+/-, and Cdh11-/- mice to identify key responses to CDH11 deletion in vivo. We showed that CDH11 influences cell behavior through its regulation of contractility and its ability to bind substrates via focal adhesions. We also show that transforming growth factor-ß1 overrides the normal relationship between CDH11 and smooth muscle α-actin to exacerbate the myofibroblast disease phenotype. This phenotypic switch is potentiated through the IL-6 signaling axis and could act as a paracrine mechanism of myofibroblast activation in neighboring aortic valve interstitial cells in a positive feedback loop. These data suggest CDH11 is an important mediator of the myofibroblast phenotype and identify several mechanisms by which it modulates cell behavior. NEW & NOTEWORTHY Cadherin-11 influences valvular interstitial cell contractility by regulating focal adhesions and inflammatory cytokine secretion. Transforming growth factor-ß1 overrides the normal balance between cadherin-11 and smooth muscle α-actin expression to promote a myofibroblast phenotype. Cadherin-11 is necessary for IL-6 and chitinase-3-like protein 1 secretion, and IL-6 promotes contractility. Targeting cadherin-11 could therapeutically influence valvular interstitial cell phenotypes in a multifaceted manner.


Assuntos
Valva Aórtica/metabolismo , Caderinas/metabolismo , Mecanotransdução Celular , Miofibroblastos/metabolismo , Actinas/metabolismo , Animais , Valva Aórtica/citologia , Caderinas/genética , Células Cultivadas , Adesões Focais/metabolismo , Interleucina-6/metabolismo , Camundongos , Ligação Proteica , Fator de Necrose Tumoral alfa/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 35(7): 1597-605, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26023079

RESUMO

OBJECTIVE: Calcific aortic valve disease (CAVD) is a significant cardiovascular disorder, and controversy exists as to whether it is primarily a dystrophic or osteogenic process in vivo. In this study, we sought to clarify the mechanism of CAVD by assessing a genetic mutation, Notch1 heterozygosity, which leads to CAVD with 100% penetrance in humans. APPROACH AND RESULTS: Murine immortalized Notch1(+/-) aortic valve interstitial cells (AVICs) were isolated and expanded in vitro. Molecular signaling of wild-type and Notch1(+/-) AVICs were compared to identify changes in pathways that have been linked to CAVD-transforming growth factor-ß1/bone morphogenetic protein, mitogen-activated protein kinase, and phosphoinositide 3-kinase/protein kinase B-and assessed for calcification potential. Additionally, AVIC mechanobiology was studied in a physiologically relevant, dynamic mechanical environment (10% cyclic strain) to investigate differences in responses between the cell types. We found that Notch1(+/-) AVICs resembled a myofibroblast-like phenotype expressing higher amounts of cadherin-11, a known mediator of dystrophic calcification, and decreased Runx2, a known osteogenic marker. We determined that cadherin-11 expression is regulated by Akt activity, and inhibition of Akt phosphorylation significantly reduced cadherin-11 expression. Moreover, in the presence of cyclic strain, Notch1(+/-) AVICs exhibited significantly upregulated phosphorylation of Akt at Ser473 and smooth muscle α-actin expression, indicative of a fully activated myofibroblast. Finally, these Notch1-mediated alterations led to enhanced dystrophic calcific nodule formation. CONCLUSIONS: This study presents novel insights in our understanding of Notch1-mediated CAVD by demonstrating that the mutation leads to AVICs that are fully activated myofibroblasts, resulting in dystrophic, but not osteogenic, calcification.


Assuntos
Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/metabolismo , Valva Aórtica/patologia , Calcinose/genética , Calcinose/metabolismo , Mecanotransdução Celular/genética , Mutação , Miofibroblastos/metabolismo , Receptor Notch1/genética , Animais , Valva Aórtica/metabolismo , Caderinas/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , MAP Quinase Quinase 2/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 33(1): 114-20, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23162011

RESUMO

OBJECTIVE: Dystrophic calcific nodule formation in vitro involves differentiation of aortic valve interstitial cells (AVICs) into a myofibroblast phenotype. Interestingly, inhibition of the kinase MAPK Erk kinase (MEK)1/2 prevents calcific nodule formation despite leading to myofibroblast activation of AVICs, indicating the presence of an additional mechanotransductive component required for calcific nodule morphogenesis. In this study, we assess the role of transforming growth factor ß1-induced cadherin-11 expression in calcific nodule formation. METHODS AND RESULTS: As shown previously, porcine AVICs treated with transforming growth factor ß1 before cyclic strain exhibit increased myofibroblast activation and significant calcific nodule formation. In addition to an increase in contractile myofibroblast markers, transforming growth factor ß1-treated AVICs exhibit significantly increased expression of cadherin-11. This expression is inhibited by the addition of U0126, a specific MEK1/2 inhibitor. The role of increased cadherin-11 is revealed through a wound assay, which demonstrates increased intercellular tension in transforming growth factor ß1-treated AVICs possessing cadherin-11. Furthermore, when small interfering RNA is used to knockdown cadherin-11, calcific nodule formation is abrogated, indicating that robust cell-cell connections are necessary in generating tension for calcific nodule morphogenesis. Finally, we demonstrate enrichment of cadherin-11 in human calcified leaflets. CONCLUSIONS: These results indicate the necessity of cadherin-11 for dystrophic calcific nodule formation, which proceeds through an Erk1/2-dependent pathway.


Assuntos
Valva Aórtica/metabolismo , Caderinas/metabolismo , Calcinose/metabolismo , Comunicação Celular , Doenças das Valvas Cardíacas/metabolismo , Miofibroblastos/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Valva Aórtica/efeitos dos fármacos , Valva Aórtica/patologia , Caderinas/genética , Calcinose/patologia , Comunicação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Doenças das Valvas Cardíacas/patologia , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Morfogênese , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/patologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Transdução de Sinais , Estresse Mecânico , Suínos , Transfecção , Fator de Crescimento Transformador beta1/metabolismo
5.
J Mol Cell Cardiol ; 53(5): 707-14, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22940605

RESUMO

Transforming growth factor-ß1 (TGF-ß1) induces myofibroblast activation of quiescent aortic valve interstitial cells (AVICs), a differentiation process implicated in calcific aortic valve disease (CAVD). The ubiquity of TGF-ß1 signaling makes it difficult to target in a tissue specific manner; however, the serotonin 2B receptor (5-HT(2B)) is highly localized to cardiopulmonary tissues and agonism of this receptor displays pro-fibrotic effects in a TGF-ß1-dependent manner. Therefore, we hypothesized that antagonism of 5-HT(2B) opposes TGF-ß1-induced pathologic differentiation of AVICs and may offer a druggable target to prevent CAVD. To test this hypothesis, we assessed the interaction of 5-HT(2B) antagonism with canonical and non-canonical TGF-ß1 pathways to inhibit TGF-ß1-induced activation of isolated porcine AVICs in vitro. Here we show that AVIC activation and subsequent calcific nodule formation is completely mitigated by 5-HT(2B) antagonism. Interestingly, 5-HT(2B) antagonism does not inhibit canonical TGF-ß1 signaling as identified by Smad3 phosphorylation and activation of a partial plasminogen activator inhibitor-1 promoter (PAI-1, a transcriptional target of Smad3), but prevents non-canonical p38 MAPK phosphorylation. It was initially suspected that 5-HT(2B) antagonism prevents Src tyrosine kinase phosphorylation; however, we found that this is not the case and time-lapse microscopy indicates that 5-HT(2B) antagonism prevents non-canonical TGF-ß1 signaling by physically arresting Src tyrosine kinase. This study demonstrates the necessity of non-canonical TGF-ß1 signaling in leading to pathologic AVIC differentiation. Moreover, we believe that the results of this study suggest 5-HT(2B) antagonism as a novel therapeutic approach for CAVD that merits further investigation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Indóis/farmacologia , Miofibroblastos/fisiologia , Piridinas/farmacologia , Receptor 5-HT2B de Serotonina/metabolismo , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Fator de Crescimento Transformador beta1/fisiologia , Ureia/análogos & derivados , Actinas/genética , Actinas/metabolismo , Animais , Doenças da Aorta/patologia , Valva Aórtica/patologia , Calcinose/patologia , Células Cultivadas , Expressão Gênica , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Fosforilação , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Transporte Proteico , Transdução de Sinais , Proteína Smad3/metabolismo , Sus scrofa , Ativação Transcricional , Ureia/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/metabolismo , Quinases da Família src/fisiologia
6.
J Biol Chem ; 285(27): 20769-79, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20430882

RESUMO

The docking protein p130Cas is a major Src substrate involved in integrin signaling and mechanotransduction. Tyrosine phosphorylation of p130Cas in focal adhesions (FAs) has been linked to enhanced cell migration, invasion, proliferation, and survival. However, the mechanism of p130Cas targeting to FAs is uncertain, and dynamic aspects of its localization have not been explored. Using live cell microscopy, we show that fluorophore-tagged p130Cas is a component of FAs throughout the FA assembly and disassembly stages, although it resides transiently in FAs with a high mobile fraction. Deletion of either the N-terminal Src homology 3 (SH3) domain or the Cas-family C-terminal homology (CCH) domain significantly impaired p130Cas FA localization, and deletion of both domains resulted in full exclusion. Focal adhesion kinase was implicated in the FA targeting function of the p130Cas SH3 domain. Consistent with their roles in FA targeting, both the SH3 and CCH domains were found necessary for p130Cas to fully undergo tyrosine phosphorylation and promote cell migration. By revealing the capacity of p130Cas to function in FAs throughout their lifetime, clarifying FA targeting mechanism, and demonstrating the functional importance of the highly conserved CCH domain, our results advance the understanding of an important aspect of integrin signaling.


Assuntos
Proteína Substrato Associada a Crk/metabolismo , Adesões Focais/metabolismo , Animais , Anticorpos Monoclonais , Movimento Celular , Proteína Substrato Associada a Crk/análise , Proteína Substrato Associada a Crk/genética , Fibroblastos/metabolismo , Genes Reporter , Variação Genética , Immunoblotting , Proteínas Luminescentes/genética , Camundongos/embriologia , Paxilina/análise , Paxilina/genética , Fosforilação , Plasmídeos , Reação em Cadeia da Polimerase , Especificidade por Substrato , Cicatrização/fisiologia , Quinases da Família src/metabolismo
7.
PLoS One ; 11(2): e0148657, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26863209

RESUMO

Serotonergic anorexigens are the primary pharmacologic risk factor associated with pulmonary arterial hypertension (PAH), and the resulting PAH is clinically indistinguishable from the heritable form of disease, associated with BMPR2 mutations. Both BMPR2 mutation and agonists to the serotonin receptor HTR2B have been shown to cause activation of SRC tyrosine kinase; conversely, antagonists to HTR2B inhibit SRC trafficking and downstream function. To test the hypothesis that a HTR2B antagonist can prevent BMRP2 mutation induced PAH by restricting aberrant SRC trafficking and downstream activity, we exposed BMPR2 mutant mice, which spontaneously develop PAH, to a HTR2B antagonist, SB204741, to block the SRC activation caused by BMPR2 mutation. SB204741 prevented the development of PAH in BMPR2 mutant mice, reduced recruitment of inflammatory cells to their lungs, and reduced muscularization of their blood vessels. By atomic force microscopy, we determined that BMPR2 mutant mice normally had a doubling of vessel stiffness, which was substantially normalized by HTR2B inhibition. SB204741 reduced SRC phosphorylation and downstream activity in BMPR2 mutant mice. Gene expression arrays indicate that the primary changes were in cytoskeletal and muscle contractility genes. These results were confirmed by gel contraction assays showing that HTR2B inhibition nearly normalizes the 400% increase in gel contraction normally seen in BMPR2 mutant smooth muscle cells. Heritable PAH results from increased SRC activation, cellular contraction, and vascular resistance, but antagonism of HTR2B prevents SRC phosphorylation, downstream activity, and PAH in BMPR2 mutant mice.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Hipertensão Pulmonar/prevenção & controle , Indóis/farmacologia , Receptor 5-HT2B de Serotonina/genética , Antagonistas da Serotonina/farmacologia , Ureia/análogos & derivados , Quinases da Família src/genética , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Movimento Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Contração Muscular/efeitos dos fármacos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Transporte Proteico , Receptor 5-HT2B de Serotonina/metabolismo , Transdução de Sinais , Ureia/farmacologia , Rigidez Vascular/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
8.
Cell Mol Bioeng ; 7(3): 446-459, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33072223

RESUMO

Fibrotic disease is a major cause of morbidity and mortality and is characterized by the transition of resident fibroblast cells into active myofibroblasts, identified by their expression of alpha smooth muscle actin. Myofibroblast differentiation is regulated by growth factor signaling and mechanical signals transduced through integrins, which converge at focal adhesion proteins (Src and FAK) and MAPK signaling, but lead to divergent outcomes. While details are known about individual pathways, little is known about their interactions. To this end, an ODE-based model of this cell signaling network was developed in parallel with in vitro experiments to analyze potential mechanisms of crosstalk and regulation of αSMA production. We found that cells lacking Src or FAK produce significantly less or more αSMA than wild type cells, respectively. Transforming growth factor beta 1 and fibroblast growth factor signal through ERK and MAPK p38 with different dynamic profiles to increase or decrease αSMA expression, respectively. Our model effectively recreated αSMA expression levels across a set of 22 experimental conditions and matched some features of transient phosphorylation of ERK and p38. These results support a potential mechanism for regulation of fibroblast differentiation: αSMA production is promoted by active p38 and Src and opposed by ERK.

9.
PLoS One ; 5(10): e13412, 2010 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-20976150

RESUMO

The docking protein p130Cas is a prominent Src substrate found in focal adhesions (FAs) and is implicated in regulating critical aspects of cell motility including FA disassembly and protrusion of the leading edge plasma membrane. To better understand how p130Cas acts to promote these events we examined requirements for established p130Cas signaling motifs including the SH3-binding site of the Src binding domain (SBD) and the tyrosine phosphorylation sites within the substrate domain (SD). Expression of wild type p130Cas in Cas -/- mouse embryo fibroblasts resulted in enhanced cell migration associated with increased leading-edge actin flux, increased rates of FA assembly/disassembly, and uninterrupted FA turnover. Variants lacking either the SD phosphorylation sites or the SBD SH3-binding motif were able to partially restore the migration response, while only a variant lacking both signaling functions was fully defective. Notably, the migration defects associated with p130Cas signaling-deficient variants correlated with longer FA lifetimes resulting from aborted FA disassembly attempts. However the SD mutational variant was fully defective in increasing actin assembly at the protruding leading edge and FA assembly/disassembly rates, indicating that SD phosphorylation is the sole p130Cas signaling function in regulating these processes. Our results provide the first quantitative evidence supporting roles for p130Cas SD tyrosine phosphorylation in promoting both leading edge actin flux and FA turnover during cell migration, while further revealing that the p130Cas SBD has a function in cell migration and sustained FA disassembly that is distinct from its known role of promoting SD tyrosine phosphorylation.


Assuntos
Movimento Celular , Proteína Substrato Associada a Crk/fisiologia , Adesões Focais , Quinases da Família src/metabolismo , Animais , Proteína Substrato Associada a Crk/genética , Proteína Substrato Associada a Crk/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Transdução de Sinais , Especificidade por Substrato , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA