Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Clin Sci (Lond) ; 129(2): 129-45, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25751394

RESUMEN

Mindin/spondin 2, an extracellular matrix (ECM) component that belongs to the thrombospondin type 1 (TSR) class of molecules, plays prominent roles in the regulation of inflammatory responses, angiogenesis and metabolic disorders. Our most recent studies indicated that mindin is largely involved in the initiation and development of cardiac and cerebrovascular diseases [Zhu et al. (2014) J. Hepatol. 60, 1046-1054; Bian et al. (2012) J. Mol. Med. 90, 895-910; Wang et al. (2013) Exp. Neurol. 247, 506-516; Yan et al. (2011) Cardiovasc. Res. 92, 85-94]. However, the regulatory functions of mindin in neointima formation remain unclear. In the present study, mindin expression was significantly down-regulated in platelet-derived growth factor-BB (PDGF-BB)-stimulated vascular smooth muscle cells (VSMCs) and wire injury-stimulated vascular tissue. Using a gain-of-function approach, overexpression of mindin in VSMCs exhibited strong anti-proliferative and anti-migratory effects on VSMCs, whereas significant suppression of intimal hyperplasia was observed in transgenic (TG) mice expressing mindin specifically in smooth muscle cells (SMCs). These mice exhibited blunted VSMC proliferation, migration and phenotypic switching. Conversely, deletion of mindin dramatically exacerbated neointima formation in a wire-injury mouse model, which was further confirmed in a balloon injury-induced vascular lesion model using a novel mindin-KO (knockout) rat strain. From a mechanistic standpoint, the AKT (Protein Kinase B)-GSK3ß (glycogen synthase kinase 3ß)/mTOR (mammalian target of rapamycin)-FOXO3A (forkhead box O)-FOXO1 signalling axis is responsible for the regulation of mindin during intimal thickening. Interestingly, an AKT inhibitor largely reversed mindin-KO-induced aggravated hyperplasia, suggesting that mindin-mediated neointima formation is AKT-dependent. Taken together, our findings demonstrate that mindin protects against vascular hyperplasia by suppression of abnormal VSMC proliferation, migration and phenotypic switching in an AKT-dependent manner. Up-regulation of mindin might represent an effective therapy for vascular-remodelling-related diseases.


Asunto(s)
Traumatismos de las Arterias Carótidas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica , Genotipo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Transducción de Señal , Factores de Tiempo
2.
J Cell Biochem ; 115(2): 349-58, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24038435

RESUMEN

The cytoplasmic signaling protein tumor necrosis factor (TNF) receptor-associated factor 5 (TRAF5), which was identified as a signal transducer for members of the TNF receptor super-family, has been implicated in several biological functions in T/B lymphocytes and the innate immune response against viral infection. However, the role of TRAF5 in cardiac hypertrophy has not been reported. In the present study, we investigated the effect of TRAF5 on the development of pathological cardiac hypertrophy induced by transthoracic aorta constriction (TAC) and further explored the underlying molecular mechanisms. Cardiac hypertrophy and function were evaluated with echocardiography, hemodynamic measurements, pathological and molecular analyses. For the first time, we found that TRAF5 deficiency substantially aggravated cardiac hypertrophy, cardiac dysfunction and fibrosis in response to pressure overload after 4 weeks of TAC compared to wild-type (WT) mice. Moreover, the mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK)-extracellular signal-regulated kinases 1/2 (ERK1/2) signaling pathway was more activated in TRAF5-deficient mice than WT mice. In conclusion, our results suggest that as an intrinsic cardioprotective factor, TRAF5 plays a crucial role in the development of cardiac hypertrophy through the negative regulation of the MEK-ERK1/2 pathway. J. Cell. Biochem. 115: 349-358, 2014. © 2013 Wiley Periodicals, Inc.


Asunto(s)
Cardiomegalia/genética , Fibrosis/genética , Transducción de Señal/genética , Factor 5 Asociado a Receptor de TNF/biosíntesis , Animales , Aorta/metabolismo , Aorta/patología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Constricción , Quinasas MAP Reguladas por Señal Extracelular/genética , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Factor 5 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 5 Asociado a Receptor de TNF/genética
3.
J Neurochem ; 126(3): 400-14, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23413803

RESUMEN

Tumor necrosis factor receptor-associated factor 5 (TRAF5) is an adaptor protein of the tumor necrosis factor (TNF) receptor superfamily and the interleukin-1 receptor/Toll-like receptor superfamily and plays important roles in regulating multiple signaling pathways. This study was conducted to investigate the role of TRAF5 in the context of brain ischemia/reperfusion (I/R) injury. Transient occlusion of the middle cerebral artery was performed on TRAF5 knockout mice (KO), neuron-specific TRAF5 transgene (TG), and the appropriate controls. Compared with the WT mice, the TRAF5 KO mice showed lower infarct volumes and better outcomes in the neurological tests. A low neuronal apoptosis level, an attenuated blood-brain barrier (BBB) disruption and an inhibited inflammatory response were exhibited in TRAF5 KO mice. TRAF5 TG mice exhibited an opposite phenotype. Moreover, the Akt/FoxO1 signaling pathway was enhanced in the ischemic brains of the TRAF5 KO mice. These results provide the first demonstration that TRAF5 is a critical mediator of I/R injury in an experimental stroke model. The Akt /FoxO1 signaling pathway probably plays an important role in the biological function of TRAF5 in this model.


Asunto(s)
Infarto Encefálico/metabolismo , Transducción de Señal/fisiología , Factor 5 Asociado a Receptor de TNF/metabolismo , Animales , Western Blotting , Infarto Encefálico/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/metabolismo
4.
Mol Cell Biochem ; 373(1-2): 53-62, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23065380

RESUMEN

Sinomenine, a pure alkaloid extract from Sinomenium acutum, has anti-inflammatory and immunoregulatory functions. This study investigated the efficiency and the signalling pathways involved in the effect of sinomenine on vascular smooth muscle cell (VSMC) dedifferentiation in response to platelet-derived growth factor (PDGF)-BB stimulation and vascular injury. VSMCs were isolated from rat aorta and preincubated with sinomenine before being stimulated with PDGF-BB. WST and BrdU incorporation assays were used to evaluate VSMC proliferation. Flow cytometric analysis was performed for testing the cell cycle progression. The cell migration of VSMCs were analysed using a Transwell system. The expression of VSMC specific genes and signalling proteins were tested by Western blot. For the animal study, C57/BL6 mice were fed either normal rodent chow diets or sinomenine chow diets that supplemented with 0.09 % sinomenine (w/w) in the normal chows for 14 days before carotid artery wire injury. PDGF-BB activated the dedifferentiation of VSMCs characterised by decreased expression of SMA, Smoothelin and SM22α. However, sinomenine treatment preserved the dedifferentiation in response to PDGF-BB. The activations of mitogen-activated protein kinase extracellular signal-regulated kinases, Akt, GSK3ß and STAT3 induced by PDGF-BB were also inhibited in sinomenine-treated VSMCs. In vivo evidence with wire-injured mice exhibited a reduction in neointimal area and an increase in smooth muscle-specific gene expression in the sinomenine-treated group. In this study, we found that sinomenine-suppressed VSMC phenotype switching induced by PDGF-BB in vitro and neointimal formation in vivo. Therefore, sinomenine is a potential candidate to be used in the treatment of vascular proliferative disease.


Asunto(s)
Desdiferenciación Celular/efectos de los fármacos , Morfinanos/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Neointima/prevención & control , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Transcripción STAT3/metabolismo
5.
J Cell Biochem ; 113(4): 1198-207, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22095643

RESUMEN

Abnormal proliferation, migration, and phenotypic modulation of vascular smooth muscle cells (VSMCs) are critical factors in neointima formation during restenosis. The purpose of this study is to determine the efficacy and possible cell signaling mechanisms of apigenin in VSMC activation induced by platelet-derived growth factor (PDGF)-BB and injury-induced neointima formation. Our data revealed a dose-dependent apigenin inhibition of PDGF-BB-induced proliferation of VSMCs by arresting cells in G0/G1-phase of the cell cycle as determined using 5-bromo-2'-deoxyuridine incorporation and flow cytometry. This was associated with the inhibition of cyclin-dependent kinase (CDK) 4,6 expression and an increase in p27Kip1 levels in PDGF-stimulated VSMCs. Moreover, apigenin was also found to regulate PDGF-induced migration and expression of smooth-muscle-specific contractile markers. Mechanistically, the PDGF-BB-induced phosphorylation of PDGF-receptor ß (PDGF-Rß), Akt/glycogen synthase kinase(GSK)3ß, extracellular signal-regulated kinase1/2 (ERK1/2), and signal transducers and activators of transcription 3 (STAT3) is negatively modulated by apigenin. For the in vivo studies using a mouse carotid arterial injury model, the administration of apigenin resulted in a significant inhibition of the neointima/media ratio and proliferating cell nuclear antigen (PCNA)-positive cells. These results demonstrate that apigenin can suppress PDGF-induced VSMC activation and neointima hyperplasia after vascular injury; these beneficial effects are probably the result of the blockade of PDGF-Rß phosphorylation and its downstream signal transduction, including the Akt/GSK-3ß, ERK1/2, and STAT3 pathways. The results suggest that apigenin may be a potential therapeutic candidate for the prevention of restenosis.


Asunto(s)
Apigenina/farmacología , Músculo Liso Vascular/crecimiento & desarrollo , Túnica Íntima/efectos de los fármacos , Animales , Becaplermina , Western Blotting , Ciclo Celular , Proliferación Celular , Células Cultivadas , Replicación del ADN , Regulación hacia Abajo/efectos de los fármacos , Inmunohistoquímica , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Túnica Íntima/crecimiento & desarrollo , Túnica Íntima/metabolismo
6.
Basic Res Cardiol ; 107(1): 239, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22202974

RESUMEN

Cellular FLICE-inhibitory protein (cFLIP) is a member of the tumour necrosis factor signalling pathway and a regulator of apoptosis, and it has a role in cardiac remodelling following myocardial infarction (MI) that remains largely uncharacterised. This study aimed to determine the function of cFLIP as a potential mediator of post-infarction cardiac remodelling. Our results show diminished cFLIP expression in failing human and murine post-infarction hearts. Genetically engineered cFLIP heterozygous (cFLIP+/-, HET) mice, cardiac-specific cFLIP-overexpressing transgenic (TG) mice and their respective wild-type (WT) and non-transgenic controls were subjected to MI by permanent ligation of their left anterior descending artery. Cardiac structure and function were assessed by echocardiography and pressure-volume loop analysis. Apoptosis, inflammation, angiogenesis, and fibrosis were evaluated in the myocardium. The HET mice showed exacerbated left ventricular (LV) contractile dysfunction, dilatation, and remodelling compared with WT mice 28 days after MI. Impaired LV function in the HET mice was associated with increases in infarct size, hypertrophy, apoptosis, inflammation, and interstitial fibrosis, and reduced capillary density. The TG mice displayed the opposite phenotype after MI. Moreover, adenovirus-mediated overexpression of cFLIP decreased LV dilatation and improved LV function and remodelling in both HET and WT mice. Further analysis of signalling events suggests that cFLIP promotes cardioprotection by interrupting JNK1/2 signalling and augmenting Akt signalling. In conclusion, our results indicate that cFLIP protects against the development of post-infarction cardiac remodelling. Thus, cFLIP gene delivery shows promise as a clinically powerful and novel therapeutic strategy for the treatment of heart failure after MI.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Insuficiencia Cardíaca/metabolismo , Infarto del Miocardio/metabolismo , Remodelación Ventricular , Animales , Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proliferación Celular , Células Endoteliales/fisiología , Fibrosis , Terapia Genética , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/inmunología , Infarto del Miocardio/mortalidad , Miocardio/patología , Neovascularización Fisiológica , Proteínas Proto-Oncogénicas c-akt/metabolismo
7.
Comput Intell Neurosci ; 2022: 3407494, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35655492

RESUMEN

Multi-intelligent decision-making has a good development at present. Based on a series of technologies such as artificial intelligence, multi-intelligent decision-making is involved in many aspects, and the country also attaches great importance to the development of such science and technology. At the same time, at present, the development of the sports industry is not balanced. Even if the country values the sports industry, it needs investment in science and technology. This paper studies the scheduling scheme of aerobics competition in sports competition. By introducing the design scheme of a multi-intelligent decision-making system and improving the MFDRL-CTDE algorithm, the similarity between the action sequence of participants in aerobics competition and the standard action sequence is obtained. Three algorithms including Markov, MFDRL-CTDE algorithm, and improved MFDRL-CTDE algorithm are used for simulation experiments, and the improved MFDRL-CTDE algorithm is more effective and stable for the decision-making of aerobics competition.


Asunto(s)
Algoritmos , Inteligencia Artificial , Simulación por Computador , Humanos
8.
Clin Chem ; 55(2): 274-84, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19056836

RESUMEN

BACKGROUND: Prospective studies have shown that C-reactive protein (CRP) is a predictor of hypertension. Because of confounding variables, a causal linkage between CRP and hypertension has not been clearly shown. We investigated whether high circulating concentrations of human CRP can induce hypertension in rats. METHODS: We administered a single intravenous injection of adeno-associated virus-green fluorescent protein (AAV-GFP) or AAV-hCRP and measured blood pressure. Using ELISA, we measured serum hCRP, serum endothelin 1 (ET-1), and urine cGMP, and we measured serum nitric oxide (NO) using the Griess method. We recorded heart rate, maximum pressure, arterial elastance, mean aortic pressure, cardiac output, and maximum rate of rise in left ventricular pressure (dP/dt max). RESULTS: A single injection of AAV-hCRP resulted in efficient and sustained hCRP expression and led to increased blood pressure 2 months after gene transfer that persisted for another 2 months. This effect was associated with decreased NO production, as demonstrated by decreased serum NO concentration and urinary cGMP excretion, and impairment of endothelial-dependent vascular relaxation. CRP transduction also increased expression of angiotensin type 1 receptor, ET-1, and endothelin type A receptor, decreased expression of angiotensin type 2 receptor and endothelial NO synthase in thoracic aortas, and increased arterial stiffness. Ex vivo studies indicated a similar detrimental effect of CRP that was reversed by the NO donor. CONCLUSION: AAV vector-mediated CRP expression resulted in hypertension mediated through reduced NO production and subsequent alteration in ET-1 and renin-angiotensin system activation. Impaired arterial elasticity may also contribute to CRP-induced hypertension. These results support a causal role for CRP in the pathogenesis of hypertension.


Asunto(s)
Adenoviridae/genética , Proteína C-Reactiva/biosíntesis , Endotelio Vascular/fisiopatología , Hipertensión/etiología , Animales , Western Blotting , Proteína C-Reactiva/análisis , Proteína C-Reactiva/genética , Proteína C-Reactiva/orina , Endotelio Vascular/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Hemodinámica/genética , Hemodinámica/fisiología , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Óxido Nítrico/biosíntesis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
BMC Pharmacol Toxicol ; 19(1): 71, 2018 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413180

RESUMEN

BACKGROUND: 3,3'-Diindolylmethane (DIM) has been extensively studied as a potential therapeutic drug with free radical scavenging, antioxidant and anti-angiogenic effects. However, whether DIM has similar effects on cardiomyocytes remains unknown. Here we evaluated DIM's influence on inflammation and apoptosis of H9C2 cardiomyocytes induced by LPS and to explore the possible mechanism of the effects. METHODS: H9C2 cells were incubated with DIM (10, 20 and 30 µM) with or without LPS for 24 h. The cytotoxicity of DIM was detected by CCK-8. The levels of tumour necrosis factor (TNF)-α and interleukin (IL)-6 were then measured using RT-qPCR and ELISA. Cell apoptosis rate and reactive oxygen species (ROS) content after DIM treatment were measured by flow cytometry. Expressions of NFκB, P-NFκB, IκBa, P-IκBa, Bax and Bcl-2 after DIM treatment were detected by western blot. The rate of NFκB nuclear translocation after DIM treatment was determined by immunocytochemical analysis. RESULTS: LPS stimulation promoted TNF-α and IL-6 mRNA expression. After treatment with various concentrations of DIM (10, 20 and 30 µM), TNF-α and IL-6 mRNA expression was clearly impaired, especially in the LPS + DIM30(µM) group. ELISA was used to measure TNF-α and IL-6 concentrations in cellular supernatant, and the result was verified to be consistent with RT-qPCR. Additionally, DIM treatment significantly blocked LPS-induced oxidative stress and inhibited LPS-induced apoptosis in H9C2 cardiomyocytes according to the results detected by flow cytometry. Moreover, compared with LPS alone, DIM significantly inhibited the LPS-induced phosphorylation of NFκB (p-NFκB) and Bax expression and increased Bcl-2 expression. CONCLUSIONS: DIM may have a protective effect for H9C2 cardiomyocytes against LPS-induced inflammatory response and apoptosis. DIM may be a new insight into the treatment of septic cardiomyopathy.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Indoles/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Interleucina-6/genética , Lipopolisacáridos , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/genética , Proteína X Asociada a bcl-2/metabolismo
10.
J Am Heart Assoc ; 6(2)2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28209562

RESUMEN

BACKGROUND: Vinexin ß is a novel adaptor protein that regulates cellular adhesion, cytoskeletal reorganization, signal transduction, and transcription; however, the exact role that vinexin ß plays in atherosclerosis remains unknown. METHODS AND RESULTS: Immunoblot analysis showed that vinexin ß expression is upregulated in the atherosclerotic lesions of both patients with coronary heart disease and hyperlipemic apolipoprotein E-deficient mice and is primarily localized in macrophages indicated by immunofluorescence staining. The high-fat diet-induced double-knockout mice exhibited lower aortic plaque burdens than apolipoprotein E-/- littermates and decreased macrophage content. Vinexin ß deficiency improved plaque stability by attenuating lipid accumulation and increasing smooth muscle cell content and collagen. Moreover, the bone marrow transplant experiment demonstrated that vinexin ß deficiency exerts atheroprotective effects in hematopoietic cells. Consistent with these changes, the mRNA expression of proinflammatory cytokines were downregulated in vinexin ß-/- apolipoprotein E-/- mice, whereas the anti-inflammatory M2 macrophage markers were upregulated. The immunohistochemical staining and in vitro experiments showed that deficiency of vinexin ß inhibited the accumulation of monocytes and the migration of macrophages induced by tumor necrosis factor α-stimulated human umbilical vein endothelial cells as well as macrophage proliferation. Finally, the inhibitory effects exerted by vinexin ß deficiency on foam cell formation, nuclear factor κB activation, and inflammatory cytokine expression were largely reversed by constitutive Akt activation, whereas the increased expression of the nuclear factor κB subset promoted by adenoviral vinexin ß was dramatically suppressed by inhibition of AKT. CONCLUSIONS: Vinexin ß deficiency attenuates atherogenesis primarily by suppressing vascular inflammation and inactivating Akt-nuclear factor κB signaling. Our data suggest that vinexin ß could be a therapeutic target for the treatment of atherosclerosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Aterosclerosis/genética , Enfermedad de la Arteria Coronaria/genética , Vasos Coronarios/metabolismo , Regulación de la Expresión Génica , Proteínas Musculares/genética , ARN Mensajero/genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Western Blotting , Células Cultivadas , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/biosíntesis , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Vinculina
11.
Cardiovasc Res ; 102(3): 469-79, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24596398

RESUMEN

AIMS: Vascular smooth muscle cell (VSMC) proliferation is central to the pathophysiology of neo-intima formation. Interferon regulatory factor 3 (IRF3) inhibits the growth of cancer cells and fibroblasts. However, the role of IRF3 in vascular neo-intima formation is unknown. We evaluated the protective role of IRF3 against neo-intima formation in mice and the underlying mechanisms. METHODS AND RESULTS: IRF3 expression was down-regulated in VSMCs after carotid wire injury in vivo, and in SMCs after platelet-derived growth factor (PDGF)-BB challenge in vitro. Global knockout of IRF3 (IRF3-KO) led to accelerated neo-intima formation and proliferation of VSMCs, whereas the opposite was seen in SMC-specific IRF3 transgenic mice. Mechanistically, we identified IRF3 as a novel regulator of peroxisome proliferator-activated receptor γ (PPARγ), a negative regulator of SMC proliferation after vascular injury. Binding of IRF3 to the AB domain of PPARγ in the nucleus of SMCs facilitated PPARγ transactivation, resulting in decreased proliferation cell nuclear antigen expression and suppressed proliferation. Overexpression of wild-type, but not truncated, IRF3 with a mutated IRF association domain (IAD) retained the ability to exert anti-proliferative effect. CONCLUSIONS: IRF3 inhibits VSMC proliferation and neo-intima formation after vascular injury through PPARγ activation.


Asunto(s)
Factor 3 Regulador del Interferón/fisiología , Neointima , Animales , Proliferación Celular , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , PPAR gamma/fisiología
12.
J Nutr Biochem ; 24(1): 62-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22784431

RESUMEN

The purpose of this study was to determine the effect and associated cell signaling mechanisms of indole-3-carbinol (I3C) on platelet-derived growth factor (PDGF)-BB-induced proliferation and migration of cultured vascular smooth muscle cells (VSMCs) and neointima formation in a carotid injury model. Our data demonstrated that I3C inhibited PDGF-BB-induced proliferation of VSMCs in a dose-dependent manner without causing cell cytotoxicity, as assessed by 5-bromo-2'-deoxyuridine incorporation and WST-1 assays. Further studies revealed that the antiproliferative effect of I3C was caused by the arrest of cells in both the G0/G1 and S phases. Moreover, I3C treatment inhibited migration of VSMCs and partly reversed the expression of smooth-muscle-specific contractile markers. We also demonstrated that I3C-induced growth inhibition was associated with an inhibition of the expression of cyclin D1 and cyclin-dependent kinase 4/6, as well as an increase in p27(Kip1) levels in PDGF-stimulated VSMCs. These beneficial effects of I3C on VSMCs appeared to be at least partly mediated by the inhibition of Akt and the subsequent activation of glycogen synthase kinase (GSK) 3ß. Furthermore, using a mouse carotid artery injury model, we found that treatment with 150 mg/kg I3C resulted in a significant reduction of the neointima/media ratio and cells positive for proliferating cell nuclear antigen. These results demonstrate that I3C can suppress the proliferation and migration of VSMCs and neointima hyperplasia after vascular injury via inhibition of the Akt/GSK3ß pathway and suggest that this might be feasible as part of a therapeutic strategy for vascular proliferative diseases.


Asunto(s)
Indoles/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Neointima/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Becaplermina , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Ratas , Ratas Sprague-Dawley , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos
13.
Int J Mol Med ; 30(5): 1034-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22922870

RESUMEN

Vascular smooth muscle cell (VSMC) proliferation plays a critical role in the development of vascular diseases. In the present study, we tested the efficacy and the mechanisms of action of gastrodin, a bioactive component of the Chinese herb Gastrodia elata Bl, in relation to platelet-derived growth factor-BB (PDGF-BB)-dependent cell proliferation and neointima formation after acute vascular injury. Cell experiments were performed with VSMCs isolated from rat aortas. WST and BrdU incorporation assays were used to evaluate VSMC proliferation. Eight-week-old C57BL/6 mice were used for the animal experiments. Gastrodin (150 mg/kg/day) was administered in the animal chow for 14 days, and the mice were subjected to wire injury of the left carotid artery. Our data demonstrated that gastrodin attenuated the VSMC proliferation induced by PDGF-BB, as assessed by WST assay and BrdU incorporation. Gastrodin influenced the S-phase entry of VSMCs and stabilised p27Kip1 expression. In addition, pre-incubation with sinomenine prior to PDGF-BB stimulation led to increased smooth muscle-specific gene expression, thereby inhibiting VSMC dedifferentiation. Gastrodin treatment also reduced the intimal area and the number of PCNA-positive cells. Furthermore, PDGF-BB-induced phosphorylation of ERK1/2, p38 MAPK, Akt and GSK3ß was suppressed by gastrodin. Our results suggest that gastrodin can inhibit VSMC proliferation and attenuate neointimal hyperplasia in response to vascular injury. Furthermore, the ERK1/2, p38 MAPK and Akt/GSK3ß signalling pathways were found to be involved in the effects of gastrodin.


Asunto(s)
Alcoholes Bencílicos/farmacología , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Glucósidos/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Neointima/prevención & control , Animales , Becaplermina , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Medicamentos Herbarios Chinos/uso terapéutico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas , Ratas Sprague-Dawley
14.
PLoS One ; 7(4): e34957, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22506059

RESUMEN

BACKGROUND: 3,3'Diindolylmethane (DIM), a natural phytochemical, has shown inhibitory effects on the growth and migration of a variety of cancer cells; however, whether DIM has similar effects on vascular smooth muscle cells (VSMCs) remains unknown. The purpose of this study was to assess the effects of DIM on the proliferation and migration of cultured VSMCs and neointima formation in a carotid injury model, as well as the related cell signaling mechanisms. METHODOLOGY/PRINCIPAL FINDINGS: DIM dose-dependently inhibited the platelet-derived growth factor (PDGF)-BB-induced proliferation of VSMCs without cell cytotoxicity. This inhibition was caused by a G0/G1 phase cell cycle arrest demonstrated by fluorescence-activated cell-sorting analysis. We also showed that DIM-induced growth inhibition was associated with the inhibition of the expression of cyclin D1 and cyclin-dependent kinase (CDK) 4/6 as well as an increase in p27(Kip1) levels in PDGF-stimulated VSMCs. Moreover, DIM was also found to modulate migration of VSMCs and smooth muscle-specific contractile marker expression. Mechanistically, DIM negatively modulated PDGF-BB-induced phosphorylation of PDGF-recptorß (PDGF-Rß) and the activities of downstream signaling molecules including Akt/glycogen synthase kinase(GSK)3ß, extracellular signal-regulated kinase1/2 (ERK1/2), and signal transducers and activators of transcription 3 (STAT3). Our in vivo studies using a mouse carotid arterial injury model revealed that treatment with 150 mg/kg DIM resulted in significant reduction of the neointima/media ratio and proliferating cell nuclear antigen (PCNA)-positive cells, without affecting apoptosis of vascular cells and reendothelialization. Infiltration of inflammatory cells was also inhibited by DIM administration. CONCLUSION: These results demonstrate that DIM can suppress the phenotypic modulation of VSMCs and neointima hyperplasia after vascular injury. These beneficial effects on VSMCs were at least partly mediated by the inhibition of PDGF-Rß and the activities of downstream signaling pathways. The results suggest that DIM has the potential to be a candidate for the prevention of restenosis.


Asunto(s)
Enfermedades de las Arterias Carótidas/tratamiento farmacológico , Indoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Neointima/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Becaplermina , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclina D1/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Fase G1/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Neointima/patología , Fosforilación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Ratas Sprague-Dawley , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA