Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Br J Nutr ; 131(10): 1777-1785, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38287709

RESUMEN

Previous studies revealed that consuming spicy food reduced mortality from CVD and lowered stroke risk. However, no studies reported the relationship between spicy food consumption, stroke types and dose­response. This study aimed to further explore the association between the frequency of spicy food intake and the risk of stroke in a large prospective cohort study. In this study, 50 174 participants aged 30­79 years were recruited. Spicy food consumption data were collected via a baseline survey questionnaire. Outcomes were incidence of any stroke, ischaemic stroke (IS) and haemorrhagic stroke (HS). Multivariable-adjusted Cox proportional hazard models estimated the association between the consumption of spicy food and incident stroke. Restricted cubic spline analysis was used to examine the dose­response relationship. During the median 10·7-year follow-up, 3967 strokes were recorded, including 3494 IS and 516 HS. Compared with those who never/rarely consumed spicy food, those who consumed spicy food monthly, 1­2 d/week and 3­5 d/week had hazard ratio (HR) of 0·914 (95 % CI 0·841, 0·995), 0·869 (95 % CI 0·758, 0·995) and 0·826 (95 % CI 0·714, 0·956) for overall stroke, respectively. For IS, the corresponding HR) were 0·909 (95 % CI 0·832, 0·994), 0·831 (95 % CI 0·718, 0·962) and 0·813 (95 % CI 0·696, 0·951), respectively. This protective effect showed a U-shaped dose­response relationship. For obese participants, consuming spicy food ≥ 3 d/week was negatively associated with the risk of IS. We found the consumption of spicy food was negatively associated with the risk of IS and had a U-shaped dose­response relationship with risk of IS. Individuals who consumed spicy food 3­5 d/week had a significantly lowest risk of IS.


Asunto(s)
Accidente Cerebrovascular Isquémico , Humanos , Persona de Mediana Edad , Femenino , Masculino , Estudios Prospectivos , Adulto , Anciano , Accidente Cerebrovascular Isquémico/prevención & control , Accidente Cerebrovascular Isquémico/epidemiología , Accidente Cerebrovascular Isquémico/etiología , Factores de Riesgo , Modelos de Riesgos Proporcionales , Dieta , Especias , Incidencia , Accidente Cerebrovascular/prevención & control , Accidente Cerebrovascular/epidemiología
2.
Phytomedicine ; 102: 154150, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35569185

RESUMEN

BACKGROUND: Thrombocytopenia (TP) remains a challenge in clinical hematology. TP may have serious consequences, such as recurrent skin and mucosal bleeding and increased risk of intracranial and internal organ hemorrhage. However, effective and safe therapeutic drugs for the long-term management of TP are still lacking. PURPOSE: This study aimed to identify more effective active compounds for TP therapy. METHODS: Liquid chromatography-mass spectrometry-nuclear magnetic resonance analysis was used to confirm the medicinal species and chemical structure of Hirsutine (HS). The proliferation of HS was examined by Cell Counting Kit (CCK-8) assay on cells lines. The effect of HS on megakaryocyte differentiation was analyzed by evaluating the expression of CD41, CD42b, and DNA ploidy via flow cytometry (FCM). The morphology of megakaryocytes and intermediate cells was observed using an optical microscope. K562 cells were then stained with Giemsa and benzidine. qRT-PCR was used to examine the mRNA expression of GATA-1, GATA-2, FOG-1, TAL-1, RUNX-1, NF-E2, and KLF-1 in K562 cells. Protein levels of the transcription factors were analyzed by western blotting. An MEK inhibitor was used to verify the relationship between the MEK/ERK signaling pathway and CD41/CD42b (FCM), FOG-1, and TAL-1. The Kunming thrombocytopenia mouse model was established by X-ray irradiation (4 Gy) and used to test HS activity and related hematopoietic organ index in vivo. Finally, computer simulations of molecular docking were used to predict the binding energies between HS-MEK and HS-ERK. RESULTS: We preliminarily identified HS by screening a plant-sourced compound library for natural compounds with megakaryocytic differentiation and maturation (MKD/MKM)-promoting activity. We found that HS not only enhanced MKD/MKM of K562 and Meg01 cells, but also suppressed the decline of peripheral platelet levels in X-ray-induced myelosuppressive mice. In addition, HS promoted MKD via activation of MEK-ERK-FOG1/TAL1 signaling, which may be the key molecular mechanism of HS action in TP treatment. Molecular docking simulations further verified that HS targets the signaling protein MEK with high-affinity. CONCLUSION: In this study, we report for the first time that hirsutine boosts MKD/MKM through the MEK/ERK/FOG1/TAL1 signaling pathway and thus represents a promising treatment option for TP.


Asunto(s)
Trombocitopenia , Trombopoyesis , Alcaloides , Animales , Diferenciación Celular , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Transducción de Señal , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Factores de Transcripción/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-29563785

RESUMEN

Background: China has high COPD rates, even among never-regular smokers. Little is known about nonrespiratory disease risks, especially vascular morbidity and mortality after developing airflow obstruction (AFO) in Chinese adults. Objective: We aimed to investigate the prospective association of prevalent AFO with major vascular morbidity and mortality. Materials and methods: In 2004-2008, a nationwide prospective cohort study recruited 512,891 men and women aged 30-79 years from 10 diverse localities across China, tracking cause-specific mortality and coded episodes of hospitalization for 9 years. Cox regression yielded adjusted HRs for vascular diseases comparing individuals with spirometry-defined prevalent AFO at baseline to those without. Results: Of 489,382 participants with no vascular disease at baseline, 6.8% had AFO, with prevalence rising steeply with age. Individuals with prevalent AFO had significantly increased vascular mortality (n=1,429, adjusted HR 1.29, 95% CI 1.21-1.36). There were also increased risks of hemorrhagic stroke (n=823, HR 1.18, 95% CI 1.09-1.27), major coronary events (n=635, HR 1.33, 95% CI 1.22-1.45), and heart failure (n=543, HR 2.19, 95% CI 1.98-2.41). For each outcome, the risk increased progressively with increasing COPD severity and persisted among never-regular smokers. Conclusion: Among adult Chinese, AFO was associated with significantly increased risks of major vascular morbidity and mortality. COPD management should be integrated with vascular disease prevention and treatment programs to improve long-term prognosis.


Asunto(s)
Obstrucción de las Vías Aéreas/epidemiología , Enfermedades Cardiovasculares/epidemiología , Pulmón/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Adulto , Anciano , Obstrucción de las Vías Aéreas/diagnóstico , Obstrucción de las Vías Aéreas/mortalidad , Obstrucción de las Vías Aéreas/fisiopatología , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/mortalidad , Enfermedades Cardiovasculares/fisiopatología , Distribución de Chi-Cuadrado , China/epidemiología , Comorbilidad , Volumen Espiratorio Forzado , Humanos , Persona de Mediana Edad , Prevalencia , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/mortalidad , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Medición de Riesgo , Factores de Riesgo , Fumar/efectos adversos , Fumar/epidemiología , Espirometría , Factores de Tiempo , Capacidad Vital
4.
J Cell Mol Med ; 20(7): 1319-28, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26991692

RESUMEN

A20 is a zinc finger protein associated with hypoxia. As chronic hypoxia is responsible for intimal hyperplasia and disordered angiogenesis of pulmonary artery, which are histological hallmarks of pulmonary artery hypertension, we intended to explore the role of A20 in angiogenesis of pulmonary artery endothelial cells (ECs). Here, we found a transient elevation of A20 expression in the lung tissues from hypoxic rats compared with normoxic controls. This rapid enhancement was mainly detected in the endothelium, and similar results were reproduced in vitro. During early hypoxia, genetic inhibition of A20 increased proliferation in pulmonary artery ECs, linking to advanced cell cycle progression as well as microtubule polymerization, and aggravated angiogenic effects including tube formation, cell migration and adhesion molecules expression. In addition, a negative feedback loop between nuclear factor-kappa B and A20 was confirmed. Our findings provide evidence for an adaptive role of A20 against pulmonary artery ECs angiogenesis via nuclear factor-kappa B activation.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Células Endoteliales/metabolismo , FN-kappa B/metabolismo , Neovascularización Fisiológica , Arteria Pulmonar/citología , Animales , Moléculas de Adhesión Celular/metabolismo , Ciclo Celular , Hipoxia de la Célula , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Retroalimentación Fisiológica , Masculino , Microtúbulos/metabolismo , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
5.
Am J Physiol Lung Cell Mol Physiol ; 309(5): L449-62, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26092993

RESUMEN

Pulmonary arterial hypertension (PAH) is a disease characterized by thickening of pulmonary artery walls, elevated pulmonary vascular resistance, pulmonary vascular thrombotic lesions, and right heart failure. Recent studies suggest that 15-lipoxygenase (15-LO)/15-hydroxyeicosatetraenoic acid (15-HETE) play an important role in PAH, acting on arterial walls. Here, we show evidence for the action of the 15-LO/15-HETE signaling in the pulmonary vascular thrombotic lesions in the experimental PAH models. Platelet deposition was augmented in rats exposed to hypoxia and Sugen 5416, which were both prevented by nordihydroguaiaretic acid (NDGA), a 15-LO inhibitor. Chronic hypoxic resulted in the platelet deposition specifically in pulmonary vasculature, which was reversed by 15-LO inhibitor. The 15-LO pathway mediated in the endothelial dysfunction induced by hypoxia in vivo. Meanwhile, 15-HETE positively regulated the generation of IL-6 and monocyte chemoattractant protein-1 (MCP-1). The coagulation and platelet activation induced by hypoxia were reversed by 15-LO inhibitor NDGA or the MCP-1 inhibitor synthesis inhibitor bindarit in rats. The 15-LO/15-HETE signaling promoted the coagulation and platelet activation, which was suppressed by MCP-1 inhibition. These results therefore suggest that 15-LO/15-HETE signaling plays a role in platelet activation and pulmonary vascular thrombosis in PAH, involving MCP-1.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensión Pulmonar/patología , Trombosis/metabolismo , Resistencia Vascular/fisiología , Animales , Araquidonato 15-Lipooxigenasa/genética , Plaquetas/patología , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Citocinas/metabolismo , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/sangre , Hipoxia/patología , Indazoles/uso terapéutico , Inhibidores de la Lipooxigenasa/uso terapéutico , Masculino , Masoprocol/uso terapéutico , Activación Plaquetaria/efectos de los fármacos , Propionatos/uso terapéutico , Arteria Pulmonar/patología , Interferencia de ARN , ARN Interferente Pequeño , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Trombosis/tratamiento farmacológico , Trombosis/etiología , Resistencia Vascular/efectos de los fármacos
6.
J Recept Signal Transduct Res ; 35(4): 329-39, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25421526

RESUMEN

Stromal cell-derived factor 1 (CXCL12) and its receptor CXC chemokine receptor 4 (CXCR4) are known to modulate hypoxia-induced pulmonary hypertension (PH) and vascular remodeling by mobilization and recruitment of progenitor cells to the pulmonary vasculature. However, little is known about CXCL12/CXCR4 regulating proliferation and cell cycle progression of pulmonary arterial smooth muscle cells (PASMCs). To determine whether CXCL12/CXCR4 regulates PASMC proliferation and the cell cycle, immunohistochemistry, Western blot, bromodeoxyuridine incorporation and cell cycle analysis were preformed in this study. Our results showed that CXCR4 was induced by hypoxia in pulmonary arteries and PASMCs of rats. Hypoxia-increased cell viability, DNA synthesis and proliferating cell nuclear antigen expression were blocked by administration of CXCR4 antagonist AMD3100, silencing CXCR4 or CXCL12. Furthermore, inhibition of CXCL12/CXCR4 suppressed cell cycle progression, decreased the number of cells in S+G2/M phase and attenuated the expression of proteins that regulate the cell cycle progression at these phases. In addition, PI3K/Akt signaling mediated CXCL12/CXCR4 regulating proliferation and cell cycle progression in PASMCs. Thus, these results indicate that blockade of CXCL12/CXCR4 inhibited PASMC proliferation and cell cycle progression in hypoxia-induced PH via PI3K/Akt signaling pathway.


Asunto(s)
Quimiocina CXCL12/antagonistas & inhibidores , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Animales , Puntos de Control del Ciclo Celular/fisiología , Hipoxia de la Célula/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/fisiología , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Masculino , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Receptores CXCR4/genética , Receptores CXCR4/fisiología , Transducción de Señal , Remodelación Vascular/fisiología
7.
J Cell Physiol ; 230(7): 1489-502, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25521840

RESUMEN

Our laboratory has previously demonstrated that 15-lipoxygenase (15-LO)/15-hydroxyeicosatetr-aenoic acid (15-HETE) is involved in hypoxic pulmonary arterial hypertension (PAH). Phenotypical alterations of vascular smooth muscle cells are considered to be an important stage in the development of PAH, whereas the underlying mechanisms and signaling systems are still unclear. Here, we determined the contribution of 15-LO/15-HETE signaling in the hypoxia-induced phenotype changes of pulmonary arterial smooth muscle cells (PASMCs). To accomplish this, cellular and molecular changes in pulmonary vascular remodeling were detected in PAH patients and rats exposed to hypoxia. We found that the hypoxia-induced alterations in PASMCs phenotypes were reversed by the inhibition of 15-LO/15-HETE or inhibition of BMP4/BMPRI. Hypoxia-induced 15-LO1/2 expression in rat PASMCs was significantly abolished by small interfering RNA targeted at BMP4. Meanwhile, BMP4/BMPRI-15-LO/15-HETE had a positive feedback mechanism. Furthermore, ERK and p38MAPK act as the downstream of the 15-LO/15-HETE-BMP4/BMPRI signaling. Our results suggest that chronic hypoxia promotes phenotypical alterations of PASMCs due to the interaction between 15-LO/15-HETE and BMP4/BMPRI. Our study reveals a novel mechanism of hypoxia-induced pulmonary vascular remodeling and suggested new therapeutic strategies for the targeting of 15-LO/15-HETE and BMP4/BMPRI in PAH treatment.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Regulación de la Expresión Génica/fisiología , Miocitos del Músculo Liso/citología , Oxígeno/metabolismo , Animales , Araquidonato 15-Lipooxigenasa/genética , Proteína Morfogenética Ósea 4/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Humanos , Ácidos Hidroxieicosatetraenoicos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Arteria Pulmonar/citología , Distribución Aleatoria , Ratas
8.
J Recept Signal Transduct Res ; 34(4): 270-82, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24853029

RESUMEN

Hypoxia activates nuclear factor of activated T cells isoforms c3 (NFATc3), a Ca(2+)-dependent transcription factor in murine pulmonary arteries (PAs), and NFATc3 has been proved to be implicated in hypoxia-induced pulmonary arterial smooth muscle cells (PASMCs) proliferation, but it remains unclear whether NFATc3 acts on the apoptosis of PASMCs, an important step in PAs remodeling. Our laboratory has demonstrated that 15-hydroxyeicosatetraenoic acid (15-HETE) is a key factor in hypoxia-induced PA remodeling and can increase PASMC intracellular Ca(2+) ([Ca(2+)](i)) in rats. It is possible that NFATc3 is related with the function of 15-HETE anti-apoptosis during hypoxia. Our results identified that NFATc3 was mainly localized in rat PASMCs and was upregulated in PAs during hypoxia-induced rat pulmonary hypertension (PH), while this effect was inhibited by administration of nordihydroguaiaretic acid (NDGA), a 15-lipoxygenase (15-LO) inhibitor. Moreover, hypoxia and exogenous 15-HETE promoted the expression and nuclear translocation of NFATc3 in PASMCs, which was inhibited by NDGA or small interfering RNA targeted to rat 15-LO1 or 15-LO2. Furthermore, endogenous 15-HETE induced by hypoxia and exogenous 15-HETE suppressed serum deprivation-induced loss of rat PASMCs survival and prevented annexin V binding, mitochondrial membrane potential depolarization, DNA nick end labeling and chromatin condensation. Although all these effects were suppressed after the cells were treated with cyclosporin A (a calcineurin/NFAT inhibitor), it aggravated the apoptosis induced by serum deprivation. Thus, all these results indicate that 15-HETE-mediated PASMCs anti-apoptosis in hypoxic PH via the Ca(2+)-NFATc3 pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Araquidonato 15-Lipooxigenasa/metabolismo , Factores de Transcripción NFATC/metabolismo , Arteria Pulmonar/metabolismo , Animales , Anexina A5/metabolismo , Calcio/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Masoprocol/administración & dosificación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Miocitos del Músculo Liso/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
9.
Zhonghua Gan Zang Bing Za Zhi ; 22(3): 223-7, 2014 Mar.
Artículo en Chino | MEDLINE | ID: mdl-24824127

RESUMEN

OBJECTIVE: To explore the role of the Rho pathway in the hepatocyte growth factor (HGF) paracrine signal-mediated bone marrow-derived mesenchymal stem cell (BMSC) promotion of apoptosis of hepatic stellate cells (HSCs). METHODS: A BMSC-HSC co-culture system was established using plates with transwell inserts. Dynamic changes in response to pretreatment with the c-met blocker PHA665752 and the Rho pathway inhibitor Y-27632 were observed under an inverted phase contrast microscope at 24, 48 and 72 h of culture. Optimal intervention concentrations of Y-27632 and PHA665752 were determined by MTT assay. Expression of alpha-smooth muscle actin in HSCs was evaluated by immunohistochemistry, and the apoptosis rate of HSCs was measured by Annexin-V-FITC/propidium iodide. RhoA protein and mRNA levels were measured by western blot and quantitative real-time PCR respectively. Concentrations of HGF and hepatocyte growth factor activator (HGFA) were quantified by enzyme-linked immunosorbent assay. Between-group differences were evaluated by one-way ANOVA with P less than 0.05 indicating significance. RESULTS: The apoptosis rates of HSCs gradually and steadily increased in a time-dependent manner. The apoptosis rate of the PHA665752 pretreated group was lowest and that of the Y-27632 pretreated group was highest, with the most robust difference occurring at the 72 h time point (P less than 0.05). The mRNA and protein expression levels of RhoA decreased in a time-dependent manner in the Y-27632 pretreated group (all time points, P less than 0.05) but the expression levels increased in a time-dependent manner in the PHA665752 pretreated group (all time points, P less than 0.05). For both the PHA665752 and the Y-27632 pretreated groups, the concentration of HGF decreased in a time-dependent manner, but the concentrations in both remained significantly higher than that in the control group at all time points examined (P less than 0.05). The concentration of HGFA increased in a time-dependent manner, and the PHA665752 pretreated group showed significantly higher levels than any of the other groups at all time points examined (P less than 0.01). CONCLUSION: BMSC promotes HSC apoptosis in a co-culture system by activating HGF and down-regulating the RhoA signaling pathway.


Asunto(s)
Apoptosis , Células de la Médula Ósea/citología , Células Estrelladas Hepáticas/citología , Células Madre Mesenquimatosas/citología , Actinas/metabolismo , Animales , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Factor de Crecimiento de Hepatocito/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal
10.
Zhonghua Gan Zang Bing Za Zhi ; 22(2): 136-41, 2014 Feb.
Artículo en Chino | MEDLINE | ID: mdl-24735597

RESUMEN

OBJECTIVE: To investigate the role of activated hepatocyte growth factor (HGF) in apoptosis of hepatic stellate cells (HSCs) and in modulating the Rho signaling pathway. METHODS: HSCs were divided into the following groups: blank control, consisting of HSCs without treatment; two treatment controls, consisting of HSCs exposed to exogenous HGF at 50 ng/ml and HSCs exposed to exogenous HGF activator (HGFA) at 70 ng/ml; three experimental groups, consisting of HSCs exposed to both exogenous HGF and HGFA, HSCs pre-incubated with the HGF inhibitor c-met at 500 ng/ml for 6 hours and then exposed to exogenous HGF and HGFA, and HSCs pre-incubated with the Rho pathway inhibitor Y-27632 at 10 ng/ml and then exposed to exogenous HGF and HGFA. Activation status of the cultured HSCs was determined by change in expression of alpha-smooth muscle actin (SMA). The optimal intervention concentration of Y-27632 was determined by MTT assay. The apoptotic status of HSCs was determined by flow cytometry. Expression of the HGF-alpha chain was detected by immunofluorescence. The expression of RhoA was evaluated by PCR (for mRNA) and by immunohistochemical staining and Western blot analysis (for protein). RESULTS: Exposure to 10 mumol/L Y-27632 led to obvious growth inhibition of HGF + HGFA-induced HSCs, compared with the other concentrations tested (P less than 0.05). HGF + HGFA induced the expression of the HGF-alpha chain in a time-dependent manner (P less than 0.01); however, the increases in expression of HGF-alpha chain induced by HGF alone and HGFA alone were not significantly different from the level in the blank controls (P more than 0.05). Exposure to HGF alone and HGFA alone led to a time-dependent increase in apoptosis (24 h, 48 h, 72 h) but exposure to HGF + HGFA led to the highest levels of apoptosis (P less than 0.05). Exposure to HGF + HGFA led to a time-dependent decrease in RhoA mRNA and protein expression (P less than 0.01). CONCLUSION: Activation of hepatocyte growth factor promotes apoptosis of hepatic stellate cells by suppressing RhoA expression and down-regulating the Rho signaling pathway.

11.
Cell Physiol Biochem ; 33(4): 1130-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24733040

RESUMEN

BACKGROUND: Tanshinone IIA inhibits the proliferation of pulmonary artery smooth muscle cells (PASMCs), but the potential mechanisms of its effects on PASMCs apoptosis remain unclear. METHODS: Rat were subjected to hypoxia for 9 days with or without Tanshinone IIA treatment. PASMCs were exposed to the conditions of 2% O2 and 93% N2 for 24 h in vitro. Hematoxylin and eosin (HE) staining was used to evaluate vascular remodeling. The Cell viability was determined using cell fluorescence staining and MTT assays, and apoptosis was assessed using flow cytometry. Protein expression was quantified by Western blotting. RESULTS: Our results showed that Tanshinone IIA treatment reduced pulmonary artery media thickening in hypoxic rats. Tanshinone IIA reduced PASMC viability in a dose-dependent manner. Additionally, Tanshinone IIA promoted PASMC apoptosis, lowered Hsp60 levels, and upregulated caspase-3 expressions under hypoxic conditions. This pro-apoptotic effect of Tanshinone IIA might be due to the reduction of the phosphorylation of JAK2/STAT3 signaling markers and the increase in the levels of the downstream target, Cx43 in PASMCs. CONCLUSION: These data suggest that Tanshinone IIA promotes PASMC apoptosis during hypoxia and reverses vascular remodeling. This effect is mediated by modulating the expression of Hsp60, caspase-3, and Cx43 via the JAK2/STAT3 signaling pathway. These results might provide a new therapeutic target to explore a novel strategy for hypoxia-induced vessel remodeling.


Asunto(s)
Abietanos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Caspasa 3/metabolismo , Hipoxia de la Célula , Células Cultivadas , Chaperonina 60/metabolismo , Conexina 43/metabolismo , Janus Quinasa 2/metabolismo , Masculino , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Arteria Pulmonar/citología , Ratas , Ratas Wistar , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos
12.
Cell Signal ; 26(7): 1476-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24657469

RESUMEN

We have established that 15-hydroxyeicosatetraenoic acid is an important factor in regulation of pulmonary vascular remodeling (PVR) associated with hypoxia-induced pulmonary hypertension (PH), which is further metabolized by 15-hydroxyprostaglandin dehydrogenase (15-PGDH) to form 15-ketoeicosatetraenoic acid (15-KETE). However, the role of 15-PGDH and 15-KETE on PH has not been identified. The purpose of this study was to investigate whether 15-PGDH/15-KETE pathway regulates hypoxia-induced PVR in PH and to characterize the underlying mechanisms. To accomplish this, Immunohistochemistry, Ultra Performance Liquid Chromatography, Western blot, bromodeoxyuridine incorporation and cell cycle analysis were preformed. Our results showed that the levels of 15-PGDH expression and endogenous 15-KETE were drastically elevated in the lungs of humans with PH and hypoxic PH rats. Hypoxia stimulated pulmonary arterial smooth muscle cell (PASMC) proliferation, which seemed to be due to the increased 15-PGDH/15-KETE. 15-PGDH/15-KETE pathway was also capable of stimulating the cell cycle progression and promoting the cell cycle-related protein expression. Furthermore, 15-KETE-promoted cell cycle progression and proliferation in PASMCs depended on protease-activated receptor 2 (PAR-2). ERK1/2 signaling was likely required for 15-PGDH/15-KETE-induced PAR-2 expression under hypoxia. Our study indicates that 15-PGDH/15-KETE stimulates the cell cycle progression and proliferation of PASMCs involving ERK1/2-mediated PAR-2 expression, and contributes to hypoxia-induced PVR.


Asunto(s)
Hipoxia de la Célula , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Receptor PAR-2/metabolismo , Remodelación Vascular/genética , Animales , Ácidos Araquidónicos/química , Ácidos Araquidónicos/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/biosíntesis , Proliferación Celular , Células Cultivadas , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/química , Ácido Eicosapentaenoico/metabolismo , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Pulmón/irrigación sanguínea , Pulmón/patología , Sistema de Señalización de MAP Quinasas , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Interferencia de ARN , ARN Interferente Pequeño , Distribución Aleatoria , Ratas , Ratas Wistar , Receptor PAR-2/biosíntesis , Receptor PAR-2/genética
13.
Int J Biochem Cell Biol ; 45(6): 1031-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23474367

RESUMEN

Hepatocellular carcinoma is a typical hypervascular tumor resulted from excessive growth of tumor cells. Previous studies have demonstrated that the lipoxygenase is considered as a potential therapeutic target and have important influence on human cancers. However, whether the 15-lipoxygenase-1 (15-LO-1)/15-hydroxyeicosatetraenoic acid (15-HETE) pathway participates in the development and progression of hepatocellular carcinoma has not been reported until now. To test the hypothesis that the 15-LO-1/15-HETE signaling regulates hepatocellular carcinoma cells growth and metastasis via the phosphoinositide-3 kinase (PI3K)/protein kinase B (Akt)/heat shock protein 90 pathway, we performed these studies. Our results showed that hepatocellular carcinoma cell lines (HepG2 and SMMC7721) apoptosis and growth arrest occurred following blockade of the 15-LO pathway with a 15-LO-1 inhibitor or siRNA, and all the effects were reversed by exogenous 15-HETE. Meanwhile, 15-HETE strengthened the expression of phosphor-Akt and heat shock protein 90, and inhibited apoptosis induced by serum deprivation via promoting the interaction of Akt with heat shock protein 90. In addition, the invasion and migration of HepG2 enhanced by 15-HETE were both attenuated by the inhibitor of Akt or heat shock protein 90. These results indicate that the 15-LO-1/15-HETE pathway prevents hepatocellular carcinoma cells from apoptosis and promotes hepatocellular carcinoma progression via a specific intracellular signaling pathway centered by the interaction of Akt with heat shock protein 90, and suggest a new therapeutic target for hepatocellular carcinoma.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Ácidos Eicosanoicos/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Apoptosis/efectos de los fármacos , Apoptosis/genética , Araquidonato 15-Lipooxigenasa/genética , Movimiento Celular/genética , Proteínas HSP90 de Choque Térmico/genética , Células Hep G2 , Humanos , Ácidos Hidroxieicosatetraenoicos/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética
14.
Hypertension ; 59(5): 1006-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22392900

RESUMEN

Chronic hypoxia is the most common cause of secondary pulmonary hypertension, for which the mechanisms are still unclear. Recent studies implicated an important role for microRNAs (miRNAs) in hypoxia-mediated responses in various cellular processes, including cell apoptosis and proliferation. Therefore, we hypothesized that these regulatory molecules might be implicated in the etiology of hypoxic pulmonary hypertension. Here we show that miRNA-328, a posttranscriptional regulator, was drastically downregulated in the pulmonary artery (PA) after a hypoxic assault. PA rings, Western blot, quantitative real-time PCR, in situ hybridization, and luciferase assay were used to investigate the role of miRNA-328 in hypoxic pulmonary hypertension. We found that hypoxia produced a significant inhibition of miRNA-328 expression, which was involved in PA vasoconstriction and remodeling. Overexpressing miRNA-328 in the transgenic mice remarkably decreased the right ventricular systolic pressure and PA wall thickness under both normoxia and hypoxia. MiRNA-328 inhibited L-type calcium channel-α1C expression through a miRNA-328 binding site within the 3' untranslational region of L-type calcium channel-α1C. The L-type calcium channel-α1C inhibition attenuated the PA response to KCl. Furthermore, miRNA-328 suppressed the insulin growth factor 1 receptor, ultimately leading to apoptosis of pulmonary arterial smooth muscle cells. The posttranscriptional repression of L-type calcium channel-α1C and insulin growth factor 1 receptor was further confirmed by luciferase reporter assay. These results showed that miRNA-328, an important protecting factor, plays a significant role in PA constriction and remodeling by regulating multiple gene targets in hypoxic pulmonary hypertension.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Hipertensión Pulmonar/metabolismo , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Modelos Animales de Enfermedad , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Ratones , Ratones Transgénicos , MicroARNs/genética , Músculo Liso Vascular/fisiopatología , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Valores de Referencia , Sensibilidad y Especificidad , Vasoconstricción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...