Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Biomed Sci ; 29(1): 25, 2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35414069

RESUMO

BACKGROUND: Abdominal aortic aneurysm (AAA) is a relatively common and often fatal condition. A major histopathological hallmark of AAA is the severe degeneration of aortic media with loss of vascular smooth muscle cells (VSMCs), which are the main source of extracellular matrix (ECM) proteins. VSMCs and ECM homeostasis are essential in maintaining structural integrity of the aorta. Cysteine-rich protein 2 (CRP2) is a VSMC-expressed protein; however, the role of CRP2 in AAA formation is unclear. METHODS: To investigate the function of CRP2 in AAA formation, mice deficient in Apoe (Apoe-/-) or both CRP2 (gene name Csrp2) and Apoe (Csrp2-/-Apoe-/-) were subjected to an angiotensin II (Ang II) infusion model of AAA formation. Aortas were harvested at different time points and histological analysis was performed. Primary VSMCs were generated from Apoe-/- and Csrp2-/-Apoe-/- mouse aortas for in vitro mechanistic studies. RESULTS: Loss of CRP2 attenuated Ang II-induced AAA incidence and severity, accompanied by preserved smooth muscle α-actin expression and reduced elastin degradation, matrix metalloproteinase 2 (MMP2) activity, deposition of collagen, particularly collagen III (Col III), aortic tensile strength, and blood pressure. CRP2 deficiency decreased the baseline MMP2 and Col III expression in VSMCs and mitigated Ang II-induced increases of MMP2 and Col III via blunting Erk1/2 signaling. Rescue experiments were performed by reintroducing CRP2 into Csrp2-/-Apoe-/- VSMCs restored Ang II-induced Erk1/2 activation, MMP2 expression and activity, and Col III levels. CONCLUSIONS: Our results indicate that in response to Ang II stimulation, CRP2 deficiency maintains aortic VSMC density, ECM homeostasis, and structural integrity through Erk1/2-Col III and MMP2 axis and reduces AAA formation. Thus, targeting CRP2 provides a potential therapeutic strategy for AAA.


Assuntos
Angiotensina II , Aneurisma da Aorta Abdominal , Angiotensina II/efeitos adversos , Angiotensina II/metabolismo , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Apolipoproteínas E/metabolismo , Colágeno/efeitos adversos , Colágeno/metabolismo , Cisteína , Modelos Animais de Doenças , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo
2.
Cell Commun Signal ; 12: 22, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24674138

RESUMO

BACKGROUND: Vascular smooth muscle cells (VSMCs) of the arterial wall play a critical role in the development of occlusive vascular diseases. Cysteine-rich protein 2 (CRP2) is a VSMC-expressed LIM-only protein, which functionally limits VSMC migration and protects against pathological vascular remodeling. The multifunctional cytokine TGFß has been implicated to play a role in the pathogenesis of atherosclerosis through numerous downstream signaling pathways. We showed previously that TGFß upregulates CRP2 expression; however, the detailed signaling mechanisms remain unclear. RESULTS: TGFß treatment of VSMCs activated both Smad2/3 and ATF2 phosphorylation. Individually knocking down Smad2/3 or ATF2 pathways with siRNA impaired the TGFß induction of CRP2, indicating that both contribute to CRP2 expression. Inhibiting TßRI kinase activity by SB431542 or TßRI knockdown abolished Smad2/3 phosphorylation but did not alter ATF2 phosphorylation, indicating while Smad2/3 phosphorylation was TßRI-dependent ATF2 phosphorylation was independent of TßRI. Inhibiting Src kinase activity by SU6656 suppressed TGFß-induced RhoA and ATF2 activation but not Smad2 phosphorylation. Blocking ROCK activity, the major downstream target of RhoA, abolished ATF2 phosphorylation and CRP2 induction but not Smad2 phosphorylation. Furthermore, JNK inhibition with SP600125 reduced TGFß-induced ATF2 (but not Smad2) phosphorylation and CRP2 protein expression while ROCK inhibition blocked JNK activation. These results indicate that downstream of TßRII, Src family kinase-RhoA-ROCK-JNK signaling pathway mediates TßRI-independent ATF2 activation. Promoter analysis revealed that the TGFß induction of CRP2 was mediated through the CRE and SBE promoter elements that were located in close proximity. CONCLUSIONS: Our results demonstrate that two signaling pathways downstream of TGFß converge on the CRE and SBE sites of the Csrp2 promoter to cooperatively control CRP2 induction in VSMCs, which represents a previously unrecognized mechanism of VSMC gene induction by TGFß.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas com Domínio LIM/metabolismo , Músculo Liso Vascular/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/farmacologia , Fator 2 Ativador da Transcrição/genética , Fator 2 Ativador da Transcrição/metabolismo , Animais , Proteínas de Transporte/genética , Células Cultivadas , Proteínas com Domínio LIM/genética , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Regiões Promotoras Genéticas , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Mol Biol Rep ; 41(11): 7033-41, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25034893

RESUMO

Vascular smooth muscle cells (VSMCs) of the arterial wall normally display a differentiated and contractile phenotype. In response to arterial injury, VSMCs switch to a synthetic phenotype, contributing to vascular remodeling. Cysteine-rich protein 2 (CRP2) is a cytoskeletal protein expressed in VSMCs and blunts VSMC migration in part by sequestering the scaffolding protein p130Cas at focal adhesions. CRP2 deficiency in mice increases neointima formation following arterial injury. The goal of this study was to use Csrp2 promoter-lacZ transgenic mice to analyze CRP2 expression during VSMC phenotypic modulation. In a neointima formation model after carotid artery cessation of blood flow, lacZ reporter activity and smooth muscle (SM) α-actin expression in the media were rapidly downregulated 4 days after carotid ligation. Fourteen days after ligation, there was a high level expression of both Csrp2 promoter activity and SM α-actin protein expression in neointimal cells. In atherosclerosis prone mice fed an atherogenic diet, Csrp2 promoter activity was detected within complex atherosclerotic lesions. Interestingly, Csrp2 promoter activity was also present in the fibrous caps of complicated atherosclerotic lesions, indicating that CRP2 might contribute to plaque stability. These findings support the concept that CRP2 contributes to the phenotypic modulation of VSMCs during vascular disease. Modulating transcription to increase CRP2 expression during vascular injury might attenuate vascular remodeling. In addition, increased CRP2 expression at the fibrous caps of advanced lesions might also serve to protect atherosclerotic plaques from rupture.


Assuntos
Aterosclerose/metabolismo , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas com Domínio LIM/metabolismo , Músculo Liso Vascular/metabolismo , Lesões do Sistema Vascular/metabolismo , Actinas/metabolismo , Animais , Artérias Carótidas/cirurgia , Proteínas de Transporte/genética , Movimento Celular/fisiologia , Primers do DNA/genética , Galactosídeos , Regulação da Expressão Gênica/genética , Genótipo , Técnicas Histológicas , Imuno-Histoquímica , Indóis , Proteínas com Domínio LIM/genética , Ligadura , Masculino , Camundongos , Camundongos Transgênicos , Neointima/fisiopatologia , Reação em Cadeia da Polimerase
4.
Arterioscler Thromb Vasc Biol ; 30(4): 835-42, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20075421

RESUMO

OBJECTIVE: An absence of cysteine-rich protein 2 (CRP2) enhances vascular smooth muscle cell (VSMC) migration and increases neointima formation after arterial injury; therefore, CRP2 plays an important role in the response to vascular injury. The goal of the present study was to elucidate the molecular mechanisms that preserve CRP2 expression in the adult vasculature and thus might serve to inhibit the response to injury. METHODS AND RESULTS: We generated a series of transgenic mice harboring potential Csrp2 regulatory regions with a lacZ reporter. We determined that the 12-kb first intron was necessary for transgene activity in adult but not in developing vasculature. Within the intron we identified a 6.3-kb region that contains 2 CArG boxes. Serum response factor preferentially bound to CArG2 box in gel mobility shift and chromatin immunoprecipitation assays; additionally, serum response factor coactivator myocardin factors activated CRP2 expression via the CArG2 box. Mutational analysis revealed that CArG2 box was important in directing lacZ expression in VSMC of adult vessels. CONCLUSIONS: Although CRP2 expression during development is independent of CArG box regulatory sites, CRP2 expression in adult VSMC requires CArG2 element within the first intron. Our results suggest that distinct mechanisms regulate CRP2 expression in VSMC that are controlled by separate embryonic and adult regulatory modules.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores Etários , Envelhecimento , Animais , Artérias/crescimento & desenvolvimento , Artérias/metabolismo , Sítios de Ligação , Células Cultivadas , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Íntrons , Proteínas com Domínio LIM , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Musculares/genética , Músculo Liso Vascular/crescimento & desenvolvimento , Mutação , Proteínas Nucleares/genética , Ratos , Sequências Reguladoras de Ácido Nucleico , Proteínas Repressoras/genética , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Transfecção
5.
Aging (Albany NY) ; 11(19): 8604-8622, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31596731

RESUMO

Cardiovascular diseases remain the leading cause of morbidity and mortality worldwide, particularly among older adults. Despite the advent of medical technology, restenosis is still an issue after interventional procedures. Tryptophan metabolite 5-methoxytryptophan (5-MTP) has recently been shown to protect against systemic inflammatory responses. This study aimed to investigate the function and mechanisms of 5-MTP in interventional procedure-induced restenosis. We found that after mouse femoral artery denudation with a guide wire, 5-MTP accelerated recovery of endothelium in the denuded area and reduced vascular leakage and intimal thickening. 5-MTP increased endothelial cell proliferation in the denuded arteries and rescued TNF-α-reduced endothelial cell proliferation and migration, likely via maintaining vascular endothelial growth factor receptor 2 activation. In contrast, 5-MTP preserved differentiated phenotype of medial vascular smooth muscle cells (VSMCs) and decreased VSMC proliferation and migration. Furthermore, 5-MTP maintained expression levels of critical transcription factors for VSMC marker gene expressions via attenuated activation of p38 MAPK and NFκB-p65. Our findings uncover a novel protective mechanism of 5-MTP in restenosis. In response to denudation injury, 5-MTP attenuates intimal hyperplasia via concerted but opposing actions on endothelial cells and VSMCs. Taken together, our results suggest that 5-MTP is a valuable therapeutic target for arterial injury-induced restenosis.


Assuntos
Reestenose Coronária , Endotélio Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Triptofano/análogos & derivados , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Reestenose Coronária/metabolismo , Reestenose Coronária/prevenção & controle , Camundongos , Fatores de Proteção , Triptofano/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Redox Biol ; 15: 51-61, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29216542

RESUMO

Heme oxygenase (HO)-1 is an inducible stress response protein and well known to protect cells and tissues against injury. Despite its important function in cytoprotection against physiological stress, the role of HO-1 in embryonic stem cell (ESC) differentiation remains largely unknown. We showed previously that induced pluripotent stem (iPS) cells that lack HO-1 are more sensitive to oxidant stress-induced cell death and more prone to lose pluripotent markers upon LIF withdrawal. To elucidate the role of HO-1 in ESC differentiation and to rule out the controversy of potential gene flaws in iPS cells, we derived and established mouse HO-1 knockout ESC lines from HO-1 knockout blastocysts. Using wild type D3 and HO-1 knockout ESCs in the 3-dimensional embryoid body (EB) differentiation model, we showed that at an early time point during EB development, an absence of HO-1 led to enhanced ROS level, concomitant with increased expressions of master mesodermal regulator brachyury and endodermal marker GATA6. In addition, critical smooth muscle cell (SMC) transcription factor serum response factor and its coactivator myocardin were enhanced. Furthermore, HO-1 deficiency increased Smad2 in ESCs and EBs, revealing a role of HO-1 in controlling Smad2 level. Smad2 not only mediates mesendoderm differentiation of mouse ESCs but also SMC development. Collectively, loss of HO-1 resulted in higher level of mesodermal and SMC regulators, leading to accelerated and enhanced SMC marker SM α-actin expression. Our results reveal a previously unrecognized function of HO-1 in regulating SMC gene expressions during ESC-EB development. More importantly, our findings may provide a novel strategy in enhancing ESC differentiation toward SMC lineage.


Assuntos
Desenvolvimento Embrionário/genética , Heme Oxigenase-1/genética , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Blastocisto/metabolismo , Diferenciação Celular/genética , Corpos Embrioides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo
7.
Oncotarget ; 7(42): 67760-67776, 2016 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-27626316

RESUMO

Abdominal aortic aneurysm (AAA) is a chronic but often fatal disease in elderly population. Heme oxygenase-1 (HO-1) is a stress response protein with antioxidative and anti-inflammatory properties. HO-1 has been shown to protect against atherogenesis and arterial intimal thickening. Emerging evidences suggest that AAA and arterial occlusive disease have distinct pathogenic mechanisms. Thus, in this study we investigated the role of HO-1 in angiotensin II-induced AAA formation in HO-1+/+apoE-/- and HO-1-/-apoE-/- mice. We found that complete loss of HO-1 increased AAA incidence and rupture rate, and drastically increased aneurysmal area and severity, accompanied with severe elastin degradation and medial degeneration. Interestingly, we often observed not only AAA but also thoracic aortic aneurysm in HO-1-/-apoE-/- mice. Furthermore, reactive oxygen species levels, vascular smooth muscle cell (VSMC) loss, macrophage infiltration, matrix metalloproteinase (MMP) activity were markedly enhanced in the aneurysmal aortic wall in HO-1-/-apoE-/- mice. In addition, HO-1-/-apoE-/- VSMCs were more susceptible to oxidant-induced cell death and macrophages from HO-1-/-apoE-/- mice had aggravated responses to angiotensin II with substantial increases in inflammatory cytokine productions and MMP9 activity. Taken together, our results demonstrate the essential roles of HO-1 in suppressing the pathogenesis of AAA. Targeting HO-1 might be a promising therapeutic strategy for AAA.


Assuntos
Anemia Hemolítica/metabolismo , Aneurisma da Aorta Abdominal/metabolismo , Transtornos do Crescimento/metabolismo , Heme Oxigenase-1/deficiência , Distúrbios do Metabolismo do Ferro/metabolismo , Anemia Hemolítica/genética , Angiotensina II , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Citocinas/metabolismo , Transtornos do Crescimento/genética , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Distúrbios do Metabolismo do Ferro/genética , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo
8.
Sci Rep ; 6: 25374, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27146795

RESUMO

5-Methoxytryptophan (5-MTP), a 5-methoxyindole metabolite of tryptophan metabolism, was recently shown to suppress inflammatory mediator-induced cancer cell proliferation and migration. However, the role of 5-MTP in vascular disease is unknown. In this study, we investigated whether 5-MTP protects against vascular remodeling following arterial injury. Measurements of serum 5-MTP levels in healthy subjects and patients with coronary artery disease (CAD) showed that serum 5-MTP concentrations were inversely correlated with CAD. To test the role of 5-MTP in occlusive vascular disease, we subjected mice to a carotid artery ligation model of neointima formation and treated mice with vehicle or 5-MTP. Compared with vehicle-treated mice, 5-MTP significantly reduced intimal thickening by 40% 4 weeks after ligation. BrdU incorporation assays revealed that 5-MTP significantly reduced VSMC proliferation both in vivo and in vitro. Furthermore, 5-MTP reduced endothelial loss and detachment, ICAM-1 and VCAM-1 expressions, and inflammatory cell infiltration in the ligated arterial wall, suggesting attenuation of endothelial dysfunction. Signaling pathway analysis indicated that 5-MTP mediated its effects predominantly via suppressing p38 MAPK signaling in endothelial and VSMCs. Our data demonstrate a novel vascular protective function of 5-MTP against arterial injury-induced intimal hyperplasia. 5-MTP might be a therapeutic target for preventing and/or treating vascular remodeling.


Assuntos
Artérias/lesões , Doença da Artéria Coronariana/sangue , Músculo Liso Vascular/efeitos dos fármacos , Neointima/tratamento farmacológico , Triptofano/análogos & derivados , Lesões do Sistema Vascular/tratamento farmacológico , Idoso , Animais , Células Cultivadas , Doença da Artéria Coronariana/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Triptofano/administração & dosagem , Triptofano/sangue , Triptofano/farmacologia , Lesões do Sistema Vascular/metabolismo
9.
Cardiovasc Res ; 100(3): 461-71, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23975851

RESUMO

AIMS: Cysteine-rich protein (CRP) 2, a member of the LIM-only CRP family that contains two LIM domains, is expressed in vascular smooth muscle cells (VSMCs) of blood vessels and functions to repress VSMC migration and vascular remodelling. The goal of this study was to define the molecular mechanisms by which CRP2 regulates VSMC migration. METHODS AND RESULTS: Transfection of VSMCs with CRP2-EGFP constructs revealed that CRP2 associated with the actin cytoskeleton. In response to chemoattractant stimulation, Csrp2 (mouse CRP2 gene symbol)-deficient (Csrp2(-/-)) VSMCs exhibited increased lamellipodia formation. Re-introduction of CRP2 abrogated the enhanced lamellipodia formation and migration of Csrp2(-/-) VSMCs following chemoattractant stimulation. Mammalian 2-hybrid and co-immunoprecipitation assays demonstrated that CRP2 interacts with p130Cas, a scaffold protein important for lamellipodia formation and cell motility. Immunofluorescence staining showed that CRP2 colocalized with phospho-p130Cas at focal adhesions (FAs)/terminal ends of stress fibres in non-migrating cells. Interestingly, in migrating cells phospho-p130Cas localized to the leading edge of lamellipodia and FAs, whereas CRP2 was restricted to FAs and stress fibres. Furthermore, we demonstrated that p130Cas expression and phosphorylation promote neointima formation following arterial injury. CONCLUSION: These studies demonstrate that CRP2 sequesters p130Cas at FAs, thereby reducing lamellipodia formation and blunting VSMC migration.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Proteínas de Transporte/metabolismo , Movimento Celular , Proteína Substrato Associada a Crk/metabolismo , Proteínas com Domínio LIM/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Proteínas de Transporte/genética , Células Cultivadas , Proteína Substrato Associada a Crk/genética , Modelos Animais de Doenças , Adesões Focais/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas com Domínio LIM/deficiência , Proteínas com Domínio LIM/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/lesões , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Neointima , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Pseudópodes/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Fibras de Estresse/metabolismo , Fatores de Tempo , Transfecção
10.
J Neurol Sci ; 332(1-2): 51-5, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23827825

RESUMO

Mutations in the 5' non-coding region of GJB1 are rarely reported in patients with Charcot-Marie-Tooth disease (CMT). We therefore aimed to assess the frequency and identities of the GJB1 5' non-coding region mutations in a cohort of CMT. We analyzed the 5' non-coding region of GJB1 (including the promoter P2 and exon 1b) in 91 unrelated CMT patients without an identified genetic cause. Two mutations, c.-529T>C, and c.-459C>T, were identified in one patient each. One polymorphism, c.-713G>A, was also identified in 53 patients and 73 of the 100 control subjects. The luciferase reporter assays showed that c.-459C>T significantly reduced the luciferase expression with or without SOX10 activation, whereas c.-529T>C impaired the expression only with SOX10 co-expression. c.-713G>A had no apparent functional effect. Mutations in the 5' non-coding region of GJB1 account for 0.8% (2 of 251) of CMT and 2.2% (2 of 91) of genetically unassigned CMT in a Taiwanese cohort. As previously demonstrated, c.-459C>T and c.-529T>C may cause CMT through compromising GJB1 expression whereas c.-713G>A is a benign variant. This study highlights the pathogenic role of the GJB1 5' non-coding region mutations in CMT, and suggests that their identification should be considered for CMT patients without commonly observed mutations.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Conexinas/genética , Mutação/genética , Regiões Promotoras Genéticas/genética , Adulto , Doença de Charcot-Marie-Tooth/fisiopatologia , Estudos de Coortes , Análise Mutacional de DNA , Estimulação Elétrica , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Condução Nervosa/genética , Fatores de Transcrição SOXE/genética , Taiwan , Transfecção , Adulto Jovem , Proteína beta-1 de Junções Comunicantes
11.
Lasers Surg Med ; 40(1): 46-54, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18220263

RESUMO

BACKGROUND AND OBJECTIVES: The purpose of this study, therefore, was to determine the mechanisms by which low-energy laser irradiation (LELI) may exert some of its angiogenic effects via the PI3 kinase/eNOS signaling pathway and induce endothelial cell migration and neovascularization, an important and necessary part of wound healing. STUDY DESIGN/MATERIALS AND METHODS: The possible molecular mechanism of helium-neon (He-Ne) laser irradiation on endothelial cells was proposed. He-Ne laser at 632.5 nm was used to stimulate human umbilical vein endothelial cell (HUVEC), and its effect on cell proliferation, nitric oxide secretion, and cell migration was determined. RESULTS: Irradiation enhanced endothelial nitric oxidase synthase (eNOS) protein expression, and irradiation of less than 0.26 J/cm(2) enhanced eNOS gene expression in HUVEC. The cell migration ability was promoted for HUVEC irradiated with 0.26 J/cm(2). This agreed with the vinculin protein expression induced by irradiation. In addition, the angiogenesis was promoted. The induced eNOS expression was inhibited by LY294002, indicating that the effect of laser on EC could be attributed to the up-regulation of eNOS expression through PI3K pathway at the cellular and molecular levels as a result of the He-Ne laser. CONCLUSIONS: The study has shown that LELI increased endothelial cell proliferation, migration, NO secretion, and identified that activation of PI3K/Akt pathway was a critical step for the elevated for eNOS expression upon LELI.


Assuntos
Movimento Celular , Proliferação de Células , Terapia com Luz de Baixa Intensidade , Óxido Nítrico Sintase Tipo III/efeitos da radiação , Fosfatidilinositol 3-Quinases/efeitos da radiação , Veias Umbilicais/citologia , Cicatrização/efeitos da radiação , Análise de Variância , Western Blotting , Células Cultivadas , Humanos , Microscopia de Fluorescência , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
12.
J Biol Chem ; 283(22): 15003-14, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18387947

RESUMO

CRP2 (cysteine-rich protein) is a vascular smooth muscle cell (VSMC)-expressed LIM-only protein. CRP2 associates with the actin cytoskeleton and interacts with transcription factors in the nucleus to mediate smooth muscle cell gene expression. Using Csrp2 (gene symbol of the mouse CRP2 gene)-deficient mice, we previously demonstrated that an absence of CRP2 enhances VSMC migration and increases neointima formation following arterial injury. Despite its importance in vascular injury, the molecular mechanisms controlling CRP2 expression in VSMC are largely unknown. Transforming growth factor beta (TGFbeta), a key factor present in the vessel wall in the early phases of arterial response to injury, plays an important role in modulating lesion formation. Because both CRP2 and TGFbeta are mediators of VSMC responses, we examined the possibility that TGFbeta might regulate CRP2 expression. TGFbeta significantly induced CRP2 mRNA and protein expression in VSMCs. Promoter analysis identified a conserved cAMP-responsive element (CRE)-like site (TAACGTCA) in the Csrp2 promoter that was critical for basal promoter activity and response to TGFbeta. Gel mobility shift assays revealed that mainly ATF2 bound to this CRE-like element, and mutation of the CRE sequences abolished binding. TGFbeta enhanced the activation of ATF2, leading to increased phospho-ATF2 levels within the DNA-protein complexes. Furthermore, ATF2-transactivated Csrp2 promoter activity and TGFbeta enhanced this activation. In addition, a phosphorylation-negative ATF2 mutant construct decreased basal and TGFbeta-mediated Csrp2 promoter activity. Our results show for the first time in VSMC that TGFbeta activates ATF2 phosphorylation and Csrp2 gene expression via a CRE promoter element.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Proteínas Musculares/biossíntese , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/biossíntese , Ativação Transcricional , Fator de Crescimento Transformador beta/metabolismo , Actinas/genética , Actinas/metabolismo , Fator 2 Ativador da Transcrição/genética , Animais , Artérias/lesões , Artérias/metabolismo , Movimento Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/metabolismo , Proteínas com Domínio LIM , Camundongos , Proteínas Musculares/genética , Mutação , Proteínas Nucleares/genética , Fosforilação , Ratos , Elementos de Resposta/genética , Ativação Transcricional/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA