Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Vascular ; 28(6): 821-828, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32486969

RESUMEN

OBJECTIVES: Cardiovascular disease (CVD) remains the primary cause of morbidity and mortality worldwide. The abnormal proliferation of vascular smooth muscle cells (VSMCs) is a key event in the pathogenesis of CVD. The functional and phenotypic changes in vascular cells are mediated by complex signaling cascades that initiate and control genetic reprogramming. Many studies have demonstrated that signal transducer and activator of transcription 3 (STAT3) regulates a diverse array of functions relevant to atherosclerosis. METHODS: In this review, we summarize the studies on the STAT3-mediated proliferation of VSMCs and subsequent CVDs such as hypertension, atherosclerosis, stroke, coronary artery disease, and myocardial infarction. Furthermore, we describe the general background of STAT3, its structure, function and regulation as well as the STAT3 signaling pathway. Finally, we highlight some potential issues and propose some solutions to these issues.Results and conclusions: STAT3 activation promotes the proliferation of VSMCs by regulating the transcription of genes. Studying the mechanism of VSMC proliferation induced by the STAT3 pathway is valuable for finding therapeutic targets for CVD.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Proliferación Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Transcripción STAT3/metabolismo , Remodelación Vascular , Animales , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Humanos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , Transducción de Señal
2.
J Cell Mol Med ; 23(6): 4196-4207, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30953402

RESUMEN

Rutaecarpine attenuates hypertensive cardiac hypertrophy in the rats with abdominal artery constriction (AAC); however, its mechanism of action remains largely unknown. Our previous study indicated that NADPH oxidase 4 (Nox4) promotes angiotensin II (Ang II)-induced cardiac hypertrophy through the pathway between reactive oxygen species (ROS) and a disintegrin and metalloproteinase-17 (ADAM17) in primary cardiomyocytes. This research aimed to determine whether the Nox4-ROS-ADAM17 pathway is involved in the protective action of rutaecarpine against hypertensive cardiac hypertrophy. AAC-induced hypertensive rats were adopted to evaluate the role of rutaecarpine in hypertensive cardiac hypertrophy. Western blotting and real-time PCR were used to detect gene expression. Rutaecarpine inhibited hypertensive cardiac hypertrophy in AAC-induced hypertensive rats. These findings were confirmed by the results of in vitro experiments that rutaecarpine significantly inhibited Ang II-induced cardiac hypertrophy in primary cardiomyocytes. Likewise, rutaecarpine significantly suppressed the Nox4-ROS-ADAM17 pathway and over-activation of extracellular signal-regulated kinase (ERK) 1/2 pathway in the left ventricle of AAC-induced hypertensive rats and primary cardiomyocytes stimulated with Ang II. The inhibition of Nox4-ROS-ADAM17 pathway and over-activation of ERK1/2 might be associated with the beneficial role of rutaecarpine in hypertensive cardiac hypertrophy, thus providing additional evidence for preventing hypertensive cardiac hypertrophy with rutaecarpine.


Asunto(s)
Proteína ADAM17/genética , Cardiomegalia/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Alcaloides Indólicos/farmacología , NADPH Oxidasa 4/genética , Quinazolinas/farmacología , Angiotensina II/genética , Animales , Aorta Abdominal/patología , Cardiomegalia/etiología , Constricción Patológica/complicaciones , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión/etiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Endogámicas Dahl , Especies Reactivas de Oxígeno/metabolismo
3.
J Cell Physiol ; 233(10): 6910-6920, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29741760

RESUMEN

Previous study suggested that the receptor component protein (RCP), one of the components of calcitonin gene-related peptide (CGRP) receptor, plays a multiple role in the cellular signal transduction. The study was designed to investigate whether or not the RCP involved in the regulation of caveolin-1/extracellular signal-regulated kinases-1 and -2 (ERK1/2) signal pathway in the vascular smooth muscle cells (VSMCs) proliferation induced by static pressure. Mouse-derived VSMCs line A10 (A10 VSMCs) was served as project in this experiment. Results showed that the A10 VSMCs viability and proliferating cell nuclear antigen (PCNA) expression which were increased by static pressure were inhibited by pretreatment of CGRP. In like manner, the expressions of the decreased-caveolin-1 and the increased-phosphorylated ERK1/2 (p-ERK1/2) induced by static pressure were significantly reversed by pretreatment of CGRP, respectively. Meanwhile, the expression of RCP was up-regulated by the static pressure. Silence of RCP gene with the small interrupt RNA (siRNA) not only significantly increased A10 VSMC proliferation but also increased the expression of p-ERK1/2 in response to static pressure. When treatment of A10 VSMCs with 120-mmHg static pressure for different time, however, the protein band of caveolin-1 and RCP was the least at time point of 10 min, but the p-ERK1/2 expression was the most maximum. In conclusion, RCP maybe involved in the static pressure-induced A10 VSMCs proliferation by regulation of caveolin-1/ERK1/2 signal pathway.


Asunto(s)
Caveolina 1/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/farmacología , Línea Celular , Proliferación Celular/fisiología , Células Cultivadas , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos del Músculo Liso/metabolismo
4.
Mediators Inflamm ; 2018: 2109352, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30046277

RESUMEN

Calcitonin gene-related peptide (CGRP) has a potent protective action on the cardiovascular system; however, little is known about the role of CGRP in angiotensin II- (Ang II-) induced inflammation of vascular smooth muscle cells (VSMCs). This study is aimed at determining the anti-inflammatory effect of CGRP in Ang II-treated VSMCs and whether a disintegrin and metalloproteinase 17 (ADAM17) modulates this protective action. Small interference RNA (siRNA) and inhibitors of CGRP, epidermal growth factor receptor (EGFR), and extracellular signal-regulated kinase 1/2 (ERK1/2) were adopted to investigate their effect on Ang II-induced inflammation in VSMCs. Here, we found that CGRP could inhibit inflammation and decrease ADAM17 expression and activation of EGFR and ERK1/2 in VSMCs stimulated with Ang II. Results of siRNA demonstrated that ADAM17 siRNA attenuated Ang II-induced inflammation and up-regulation of activities of EGFR and ERK1/2 in VSMCs. Furthermore, the EGFR-ERK1/2 pathway promoted Ang II-induced VSMC inflammation. In summary, these findings identify the anti-inflammatory effect of CGRP in VSMCs stimulated by Ang II and suggest that ADAM17 is involved in the protective effect of CGRP against Ang II-induced inflammation via the EGFR-ERK1/2 pathway in VSMCs.


Asunto(s)
Proteína ADAM17/metabolismo , Angiotensina II/efectos adversos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Inflamación/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Línea Celular , Receptores ErbB/metabolismo , Inflamación/inducido químicamente , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/citología , ARN Interferente Pequeño/metabolismo , Ratas , Sistema Renina-Angiotensina
5.
Cell Physiol Biochem ; 42(4): 1603-1613, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28738389

RESUMEN

BACKGROUND/AIMS: The enhanced proliferation of pulmonary arterial smooth muscle cells (PASMCs) is a central pathological component in pulmonary arterial hypertension (PAH). Both the Warburg effect and platelet-derived growth factor (PDGF) are involved in the proliferation of PASMCs. However, the mechanism underlying the crosstalk between the Warburg effect and PDGF during PASMC proliferation is still unknown. We hypothesized that PDGF promotes the Warburg effect via activating the phosphatidylinositol 3-kinase (PI3K) signaling pathway and hypoxia-inducible factor 1-α (HIF-1α) in proliferative PASMCs. METHODS: PASMCs were derived from pulmonary arteries of SD rats; cell viability, the presence of metabolites, and metabolic enzyme activities assay were determined by MTT assays, kit assays and western blot analysis, respectively. RESULTS: PDGF promoted PASMC proliferation in a dose- and time-dependent manner, accompanied by an enhanced Warburg effect. Treatment with PDGFR antagonists, Warburg effect inhibitor and PDK1 inhibitor significantly inhibited PI3K signaling activation, HIF-1α expression and PASMC proliferation induced by PDGF, respectively. Furthermore, treatment with PI3K signaling pathway inhibitors remarkably suppressed PDGF-induced PASMC proliferation and the Warburg effect. CONCLUSION: microplate reader (Biotek, Winooski The Warburg effect plays a critical role in PDGF-induced PASMC proliferation and is mediated by activation of the PI3K signaling pathway and HIF-1α.


Asunto(s)
Glucólisis/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Miocitos del Músculo Liso/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética , Animales , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Glucólisis/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
6.
Acta Biochim Biophys Sin (Shanghai) ; 47(12): 969-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26607438

RESUMEN

Apelin is highly expressed in rat left ventricular hypertrophy Sprague Dawley rat models, and it plays a crucial role in the cardiovascular system. The aim this study was to clarify whether apelin-13 promotes hypertrophy in H9c2 rat cardiomyocytes and to investigate its underlying mechanism. The cardiomyocyte hypertrophy was observed by measuring the diameter, volume, and protein content of H9c2 cells. The activation of autophagy was evaluated by observing the morphology of autophagosomes by transmission electron microscopy, observing the subcellular localization of LC3 by light microscopy, and detecting the membrane-associated form of LC3 by western blot analysis. The phosphatidylinositol 3-kinase (PI3K) signaling pathway was identified and the proteins expression was detected using western blot analysis. The results revealed that apelin-13 increased the diameter, volume, and protein content of H9c2 cells and promoted the phosphorylation of PI3K, Akt, ERK1/2, and p70S6K. Apelin-13 activated the PI3K-Akt-ERK1/2-p70S6K pathway. PI3K inhibitor LY294002, Akt inhibitor 1701-1, ERK1/2 inhibitor PD98059 attenuated the increase of the cell diameter, volume, protein content induced by apelin-13. Apelin-13 increased the autophagosomes and up-regulated the expressions of beclin 1 and LC3-II/I both transiently and stably. The autophagy inhibitor 3MA ameliorated the increase of cell diameter, volume, and protein content that were induced by apelin-13. These results suggested that apelin-13 promotes H9c2 rat cardiomyocyte hypertrophy via PI3K-Akt-ERK1/2-p70S6K and PI3K-induced autophagy.


Asunto(s)
Autofagia , Cardiomegalia/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Miocitos Cardíacos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Apelina , Línea Celular , Cromonas/química , Inhibidores Enzimáticos/química , Flavonoides/química , Regulación Enzimológica de la Expresión Génica , Morfolinas/química , Ratas
7.
Sheng Li Xue Bao ; 67(2): 193-200, 2015 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-25896050

RESUMEN

Angiotensin II (Ang II) and calcitonin gene-related peptide (CGRP) play important roles in vascular injury and protection. In order to determine the role of CGRP receptor component protein (RCP) in signal transduction whereby CGRP and Ang II mediate the expression of vascular peroxidase-1 (VPO1) in vascular smooth muscle cell (VSMC), mouse derived A10 vascular smooth muscle cell line (A10VSMC) was cultured with CGRP or/and Ang II in vitro. RCP-specific small interference RNA (siRNA-RCP) was used to silence oligonucleotide sequence. Western blot and RT-PCR were used to determine the protein and mRNA expressions of RCP and VPO1, respectively. The results showed that the expressions of RCP and VPO1 were increased in the presence of CGRP or Ang II in the quiescent A10VSMC. But the protein expressions of RCP and VPO1 induced by Ang II were decreased by pretreatment of CGRP (P < 0.05). The expressions of VPO1 were decreased in all the groups treated with siRNA-RCP, compared with those of wide-type counterparts. Meanwhile, the expression of VPO1 was significantly induced by CGRP but not Ang II in the siRNA-RCP treated A10VSMCs. Ang II in combination with CGRP increased the protein expression of VPO1 in the siRNA-RCP-transfected cells, compared with Ang II alone, and this effect could be abolished by catalase. The results suggest that RCP may play an important role in the integration of signal transduction whereby CGRP and Ang II receptors jointly regulate VPO1 expression in VSMC.


Asunto(s)
Angiotensina II/farmacología , Péptido Relacionado con Gen de Calcitonina/farmacología , Miocitos del Músculo Liso/metabolismo , Peroxidasas/metabolismo , Animales , Ratones , Músculo Liso Vascular/citología , ARN Interferente Pequeño , Transducción de Señal
8.
Acta Biochim Biophys Sin (Shanghai) ; 46(8): 699-708, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24966188

RESUMEN

This study is designed to investigate whether APJ receptor acts as a sensor in static pressure-induced cardiomyocyte hypertrophy and to investigate the mechanism of PI3K-autophagy pathway. The left ventricular hypertrophy rat model was established by coarctation of abdominal aorta. H9c2 rat cardiomyocytes were cultured in the presence of static pressure which was given by a custom-made pressure incubator. The results revealed that the expression of apelin/APJ system, PI3K, Akt and their phosphorylation were significantly increased in the operation group. Static pressure up-regulated the APJ expression, PI3K phosphorylation, Akt phosphorylation, LC3-II/I and beclin-1 expression in cardiomyocytes. APJ shRNA pGPU6/Neo-rat-399, PI3K inhibitor LY294002, Akt inhibitor 1701-1 blocked the up-regulation of APJ, PI3K phosphorylation, Akt phosphorylation, LC3-II/I and beclin-1 expression, respectively. Moreover, static pressure increased the diameter, volume, protein content of cells, and these could be reversed when the cells were treated with pGPU6/Neo-rat-399, LY294002, and autophagy inhibitor 3-methyladenine, respectively. These results suggested that static pressure up-regulates APJ expression to promote cardiomyocyte hypertrophy by a PI3K-autophagy pathway.


Asunto(s)
Autofagia , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Animales , Receptores de Apelina , Secuencia de Bases , Línea Celular , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Miocitos Cardíacos/enzimología , Ratas , Ratas Sprague-Dawley
9.
Acta Biochim Biophys Sin (Shanghai) ; 46(2): 100-11, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24374773

RESUMEN

The aim of this study was to investigate the role of apelin in the cell proliferation and autophagy of lung adenocarcinoma. The over-expression of APJ in lung adenocarcinoma was detected by immunohistochemistry, while plasma apelin level in lung cancer patients was measured by enzyme-linked immunosorbent assay. Our findings revealed that apelin-13 significantly increased the phosphorylation of ERK1/2, the expression of cyclin D1, microtubule-associated protein 1 light chain 3A/B (LC3A/B), and beclin1, and confirmed that apelin-13 promoted A549 cell proliferation and induced A549 cell autophagy via ERK1/2 signaling. Moreover, there are pores on the surface of human lung adenocarcinoma cell line A549 and apelin-13 causes cell surface smooth and glossy as observed under atomic force microscopy. These results suggested that ERK1/2 signaling pathway mediates apelin-13-induced lung adenocarcinoma cell proliferation and autophagy. Under our experimental condition, autophagy associated with 3-methyladenine was not involved in cell proliferation.


Asunto(s)
Adenocarcinoma/fisiopatología , Autofagia/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Neoplasias Pulmonares/fisiopatología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Proteínas Reguladoras de la Apoptosis/biosíntesis , Beclina-1 , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1 , Flavonoides/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/fisiología , Neoplasias Pulmonares/patología , Proteínas de la Membrana/biosíntesis , Proteínas Asociadas a Microtúbulos/biosíntesis , Fosforilación/efectos de los fármacos , Transducción de Señal
10.
Nat Commun ; 15(1): 2271, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480731

RESUMEN

T cell receptor (TCR)-engineered T cell therapy is a promising potential treatment for solid tumors, with preliminary efficacy demonstrated in clinical trials. However, obtaining clinically effective TCR molecules remains a major challenge. We have developed a strategy for cloning tumor-specific TCRs from long-term surviving patients who have responded to immunotherapy. Here, we report the identification of a TCR (10F04), which is human leukocyte antigen (HLA)-DRA/DRB1*09:01 restricted and human papillomavirus type 18 (HPV18) E784-98 specific, from a multiple antigens stimulating cellular therapy (MASCT) benefited metastatic cervical cancer patient. Upon transduction into human T cells, the 10F04 TCR demonstrated robust antitumor activity in both in vitro and in vivo models. Notably, the TCR effectively redirected both CD4+ and CD8+ T cells to specifically recognize tumor cells and induced multiple cytokine secretion along with durable antitumor activity and outstanding safety profiles. As a result, this TCR is currently being investigated in a phase I clinical trial for treating HPV18-positive cancers. This study provides an approach for developing safe and effective TCR-T therapies, while underscoring the potential of HLA class II-restricted TCR-T therapy as a cancer treatment.


Asunto(s)
Papillomavirus Humano 18 , Neoplasias del Cuello Uterino , Femenino , Humanos , Ratones , Animales , Papillomavirus Humano 18/metabolismo , Linfocitos T CD8-positivos , Receptores de Antígenos de Linfocitos T/metabolismo , Neoplasias del Cuello Uterino/terapia , Antígenos HLA
11.
Yao Xue Xue Bao ; 48(1): 59-65, 2013 Jan.
Artículo en Zh | MEDLINE | ID: mdl-23600142

RESUMEN

To investigate the effect of losartan on the axis of prolylcarboxypeptidase (PRCP)--kallikrein of the two-kidney, one-clipped (2K1C) hypertensives rats, and explore the novel protection mechanism of losartan on the kidney. Sprague-Dawley (SD) rats were used to develop the 2K1C hypertensive rats. Then, the rats were treated with prazosin (5 mg x kg(-1) x d(-1)) or losartan (5, 15 and 45 mg x kg(-1) x d(-1)) or vehicle, separately. At the same time, the blood pressures were observed. After treated for four weeks, the ratio of right kidney weight and body weight, the change of glomerular morphology, and K+, Na+, creatinine and blood urea nitrogen (BUN) of the serum were used for evaluation of kidney. The expressions of PRCP mRNA in the kidneys were determined by RT-PCR. The protein levels of PRCP, tissue kallikrein, plasma kallikrein, TGF-beta1 in kidney or plasma were measured by Western blotting. Results showed that the changes of body weight and kidney weight ratio, glomerular fibrosis degree and the biochemistrical index of serum induced by hypertension were relieved when the hypertensive rats treated with losartan for four weeks. Meanwhile, treatment of losartan also significantly decreased expression of TGF-beta1 and increased expressions of PRCP, plasma kallikrein and tissue kallikrein. The protective effects of losartan on the kidney of 2K1C hypertensive rats are activation of the axis of PRCP-kallikrein and reducing the expression of TGF-beta1.


Asunto(s)
Antihipertensivos/farmacología , Carboxipeptidasas/metabolismo , Hipertensión Renovascular , Calicreínas/metabolismo , Riñón , Losartán/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Carboxipeptidasas/genética , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/patología , Hipertensión Renovascular/fisiopatología , Calicreínas/sangre , Riñón/metabolismo , Riñón/patología , Glomérulos Renales/patología , Masculino , Tamaño de los Órganos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/metabolismo
12.
Yao Xue Xue Bao ; 48(11): 1657-64, 2013 Nov.
Artículo en Zh | MEDLINE | ID: mdl-24475702

RESUMEN

This study is to investigate the impairment and possible mechanism of endothelium-dependent relaxation of mice mesenteric arteries induced by mmLDL. Wire myography was employed to examine endothelial function of mesenteric arteries. Ultramicrostructure of mesenteric vascular beds were detected by transmission electron microscope. The results showed that endothelium cell edema and peeling, vascular elastic membrane fracture traces in mmLDL group. Endothelium-dependent relaxation was decreased in a time-dependent and dose-dependent manner by using mmLDL, compared with normal arteries. In endothelium-derived hyperpolarizing factor (EDHF)-mediated relaxation, the Rmax and pIC50 were decreased from (63 +/- 5) % and 6.42 +/- 0.09 of normal saline control to (31 +/- 3) % and 5.67 +/- 0.07 in mmLDL group (P < 0.001, P < 0.001), respectively. In nitric oxide (NO)-mediated relaxation, the Rmax and pIC50 were decreased from (45 +/- 4) % and 5.93 +/- 0.08 in normal saline control to (32 +/- 4) % and 5.43 +/- 0.11 in mmLDL group (P < 0.05, P < 0.01), respectively. There is no significant alteration of prostacyclin I2 (PGI2) pathway between these two groups. In conclusion, mmLDL induced the impairment of the ultramicrostructure of mesenteric vascular endothelium cell as well as the endothelium-dependent relaxation. The latter includes the dysfunction of NO- and EDHF pathway mediated endothelium-dependent relaxation.


Asunto(s)
Lipoproteínas LDL/farmacología , Arterias Mesentéricas/fisiología , Vasodilatación/efectos de los fármacos , Animales , Factores Biológicos/antagonistas & inhibidores , Factores Biológicos/fisiología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/ultraestructura , Epoprostenol/antagonistas & inhibidores , Epoprostenol/fisiología , Femenino , Lipoproteínas LDL/administración & dosificación , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/ultraestructura , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Transmisión , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/fisiología
13.
Int J Biol Macromol ; 223(Pt A): 290-306, 2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36347370

RESUMEN

Nanoparticles (NPs)-based on various ionic polysaccharides, including chitosan, hyaluronic acid, and alginate have been frequently summarized for controlled release applications, however, most of the published reviews, to our knowledge, focused on the delivery of a single therapeutic agent. A comprehensive summarization of the co-delivery of multiple therapeutic agents by the ionic polysaccharides-based NPs, especially on the optimization of the polysaccharide structure for overcoming various extracellular and intracellular barriers toward maximized synergistic effects, to our knowledge, has been rarely explored so far. For this purpose, the strategies used for overcoming various extracellular and intracellular barriers in vivo were introduced first to provide guidance for the rational design of ionic polysaccharides-based NPs with desired features, including long-term circulation, enhanced cellular internalization, controllable drug/gene release, endosomal escape and improved nucleus localization. Next, four preparation strategies were summarized including three physical methods of polyelectrolyte complexation, ionic crosslinking, and self-assembly and a chemical conjugation approach. The challenges and future trends of this rapidly developing field were finally discussed in the concluding remarks. The important guidelines on the rational design of ionic polysaccharides-based NPs for maximized synergistic efficiency drawn in this review will promote the future generation and clinical translation of polysaccharides-based NPs for cancer therapy.


Asunto(s)
Quitosano , Nanopartículas , Quitosano/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Polisacáridos/química , Portadores de Fármacos/química , Terapia Genética
14.
Clin Chim Acta ; 513: 25-33, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33279502

RESUMEN

Caveolin-1 is considered an important pathophysiological factor in atherosclerosis development. Previous studies indicate that caveolin-1 exhibits a pathogenic capacity in atherosclerosis via the regulation of membrane trafficking, cholesterol metabolism and cellular signal transduction. Accumulating evidence shows that autophagy activation influences the progression and development of atherosclerosis in multiple ways, including cholesterol metabolism, inflammatory responses and lipid transcytosis. However, how caveolin-1 is involved in autophagy activation in atherosclerosis remains unclear, and the precise mechanisms of caveolin-1 on autophagic flux in atherosclerosis need to be further investigated. Clarifying the roles and mechanisms of caveolin-1 in the regulation of autophagy activation is of great importance, contributing to the ability to manipulate caveolin-1 as a novel therapeutic approach for atherosclerosis. In this review, we summarize the understanding of the molecular structure, biological roles and biochemical functions of caveolin-1 to date and discuss the roles and mechanisms of caveolin-1 in autophagy activation. The emphasis on the potential of caveolin-1 to be a novel therapeutic target in atherosclerosis and understanding its precise functions and exact mechanisms in autophagic flux will provide evidence for future experimental research and aid in the development of novel therapeutic strategies for atherosclerosis.


Asunto(s)
Aterosclerosis , Caveolina 1 , Aterosclerosis/tratamiento farmacológico , Autofagia , Caveolina 1/metabolismo , Humanos , Metabolismo de los Lípidos
15.
Front Physiol ; 12: 656139, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897463

RESUMEN

Pulmonary hypertension (PH) is a critical and dangerous disease in cardiovascular system. Pulmonary vascular remodeling is an important pathophysiological mechanism for the development of pulmonary arterial hypertension. Pulmonary artery smooth muscle cell (PASMC) proliferation, hypertrophy, and enhancing secretory activity are the main causes of pulmonary vascular remodeling. Previous studies have proven that various active substances and inflammatory factors, such as interleukin 6 (IL-6), IL-8, chemotactic factor for monocyte 1, etc., are involved in pulmonary vascular remodeling in PH. However, the underlying mechanisms of these active substances to promote the PASMC proliferation remain to be elucidated. In our study, we demonstrated that PASMC senescence, as a physiopathologic mechanism, played an essential role in hypoxia-induced PASMC proliferation. In the progression of PH, senescence PASMCs could contribute to PASMC proliferation via increasing the expression of paracrine IL-6 (senescence-associated secretory phenotype). In addition, we found that activated mTOR/S6K1 pathway can promote PASMC senescence and elevate hypoxia-induced PASMC proliferation. Further study revealed that the activation of mTOR/S6K1 pathway was responsible for senescence PASMCs inducing PASMC proliferation via paracrine IL-6. Targeted inhibition of PASMC senescence could effectively suppress PASMC proliferation and relieve pulmonary vascular remodeling in PH, indicating a potential for the exploration of novel anti-PH strategies.

16.
Acta Biochim Biophys Sin (Shanghai) ; 42(6): 396-402, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20539939

RESUMEN

Vascular smooth muscle cells (VSMCs) were prepared from thoracic aortas of male Sprague-Dawley rats by the explant method to observe VSMC proliferation via phosphoinositide 3 kinase (PI3K)/Akt signaling transduction pathway induced by apelin-13. Expression of PI3K, phospho-PI3K, phospho-Akt, ERK1/2, phospho-ERK1/2 and cyclin D1 was detected by western blot analysis. Results showed that apelin-13 promoted the expression of phospho-PI3K and phospho-Akt in dose- and timedependent manner. PI3K inhibitor LY294002 significantly decreased the expression of phospho-PI3K, phospho-Akt, phospho-ERK1/2, and cyclin D1 induced by apelin-13. The Akt inhibitor 1701-1 significantly diminished the expression of phospho-Akt, phospho-ERK1/2, and cyclin D1 stimulated by apelin-13. MTT assay results showed that PI3K inhibitor LY294002 and Akt inhibitor 1701-1 significantly inhibited the VSMC proliferation induced by apelin-13. Apelin-13 promoted VSMC proliferation through PI3K/Akt signaling transduction pathway.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/farmacología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Aorta Torácica/citología , Receptores de Apelina , Western Blotting , Bovinos , Proliferación Celular , Cromonas/farmacología , Ciclina D1/metabolismo , Masculino , Morfolinas/farmacología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
17.
Acta Biochim Biophys Sin (Shanghai) ; 42(6): 403-9, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20539940

RESUMEN

To investigate whether apelin-13 induced THP-1 monocytes (MCs) adhesion to ECV304 human umbilical vein endothelial cells (HUVECs) via 14-3-3 signaling transduction pathway and the potential novel physiological function and signaling transduction pathway of apelin-APJ, HUVECs ECV304 were cultured in DMEM and MCs THP-1 were cultured in RPMI 1640 medium. Monocyte adhesion and the expression of vascular cell adhesion molecule-1 (VCAM-1) and 14-3-3 were measured with monocyte adhesion assay and western blot analysis. Data showed that apelin-13 increased adhesion of MCs to HUVECs in a concentration- and time-dependent manner, which reached their peaks at 1 mM and 12 h, respectively. Similarly, apelin-13 induced the expression of HUVECs adhesion molecule, VCAM-1, in a concentration- and time-dependent manner, reached their peaks at 1 microM and 12 h, respectively. Apelin-13 induced the expression of 14-3-3 in a concentration- and timedependent manner, which reached their peaks at 1 mM and 5 min, respectively. Furthermore, the potent 14-3-3 inhibitor difopein significantly reduced the expression of 14-3-3 and VCAM-1 in apelin-13 stimulated HUVECs, and difopein significantly inhibited the effect of apelin-13 on induction of MCs adhesion to HUVECs. These data suggested that 14-3-3 mediated the induction of adhesion of MCs to HUVECs by Apelin-13.


Asunto(s)
Proteínas 14-3-3/metabolismo , Endotelio Vascular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Monocitos/fisiología , Proteínas 14-3-3/biosíntesis , Adhesión Celular/efectos de los fármacos , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Femenino , Humanos , Embarazo , Proteínas/farmacología , Transducción de Señal , Venas Umbilicales/citología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
Clin Exp Pharmacol Physiol ; 36(3): 319-24, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19018804

RESUMEN

1. Previous studies indicate that rutaecarpine blocks increases in blood pressure and inhibits vascular hypertrophy in experimentally hypertensive rats. The aim of the present study was to determine whether the effects of rutaecarpine are related to activation of prolylcarboxypeptidase (PRCP). 2. Renovascular hypertensive rats (Goldblatt two-kidney, one-clip (2K1C)) were developed using male Sprague-Dawley rats. Chronic treatment with rutaecarpine (10 or 40 mg/kg per day) or losartan (20 mg/kg per day) for 4 weeks to the hypertensive rats caused a sustained dose-dependent attenuation of increases in blood pressure, increased lumen diameter and decreased media thickness, which was accompanied by a similar reduction in the media cross-sectional area : lumen area ratio in mesenteric arteries compared with untreated hypertensive rats. 3. Angiotensin (Ang) II expression was significantly increased in mesenteric arteries of hypertensive rats compared with sham-operated rats. No significant differences in plasma AngII levels were observed between untreated hypertensive and sham-operated rats. Hypertensive rats treated with high-dose rutaecarpine had significantly decreased Ang II levels in both the plasma and mesenteric arteries. 4. Expression of PRCP protein or kallikrein mRNA was significantly inhibited in the right kidneys and mesenteric arteries of hypertensive rats. However, expression of PRCP protein and kallikrein mRNA was significantly increased after treatment with rutaecarpine or losartan (20 mg/kg per day). 5. The data suggest that the repression of increases in systolic blood pressure and reversal of mesenteric artery remodelling by rutaecarpine may be related to increased expression of PRCP in the circulation and small arteries in 2K1C hypertensive rats.


Asunto(s)
Antihipertensivos/farmacología , Carboxipeptidasas/metabolismo , Hipertensión Renovascular/tratamiento farmacológico , Alcaloides Indólicos/farmacología , Arterias Mesentéricas/efectos de los fármacos , Quinazolinas/farmacología , Angiotensina II/sangre , Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Carboxipeptidasas/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipertensión Renovascular/enzimología , Hipertensión Renovascular/patología , Hipertrofia , Calicreínas/metabolismo , Losartán/farmacología , Masculino , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
20.
Front Biosci ; 13: 3786-92, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508473

RESUMEN

Apelin is the endogenous ligand of the G protein-coupled receptor, APJ. Vascular smooth muscle cells express both apelin and APJ, which are important regulatory factors in the cardiovascular and nervous systems. Importantly, APJ is also involved in the pathogenesis if HIV-1 infection. We investigated whether vascular smooth muscle cell proliferation was regulated through an apelin-pERK1/2-cyclin D1 signal transduction pathway. Apelin-13 significantly stimulated vascular smooth muscle cell proliferation and increased cell cycle progression. Apelin-13 a decreased the proportion of cell in the G0/G1 phase while increasing the number of cells in S phase. Apelin-13 also increased the levels of cyclin D1, cyclin E and pERK1/2. Treatment of cells with the MEK inhibitor PD98059 attenuated the apelin-3-induced pERK1/2 activation. Similarly, treatment with PD98059 partially diminished the apelin-13-induced expression of cyclin D1 and vascular smooth muscle cell proliferation. Taken together, these data established that apelin-13 stimulates vascular smooth muscle cell proliferation by promoting the G1-S phase transition, and that this effect is mediated in part by an apelin-pERK1/2-cyclin D1 signal cascade.


Asunto(s)
Proteínas Portadoras/farmacología , División Celular/efectos de los fármacos , Ciclina D1/fisiología , Músculo Liso Vascular/citología , Animales , Aorta Torácica/citología , Apelina , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Humanos , Infecciones/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA