Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Cell Mol Med ; 24(14): 8018-8030, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32468734

RESUMO

Human urinary induced pluripotent stem cells (hUiPSCs) produced from exfoliated renal epithelial cells present in urine may provide a non-invasive source of endothelial progenitors for the treatment of ischaemic diseases. However, their differentiation efficiency is unsatisfactory and the underlying mechanism of differentiation is still unknown. Gremlin1 (GREM1) is an important gene involved in cell differentiation. Therefore, we tried to elucidate the roles of GREM1 during the differentiation and expansion of endothelial progenitors. HUiPSCs were induced into endothelial progenitors by three stages. After differentiation, GREM1 was obviously increased in hUiPSC-induced endothelial progenitors (hUiPSC-EPs). RNA interference (RNAi) was used to silence GREM1 expression in three stages, respectively. We demonstrated a stage-specific effect of GREM1 in decreasing hUiPSC-EP differentiation in the mesoderm induction stage (Stage 1), while increasing differentiation in the endothelial progenitors' induction stage (Stage 2) and expansion stage (Stage 3). Exogenous addition of GREM1 recombinant protein in the endothelial progenitors' expansion stage (Stage 3) promoted the expansion of hUiPSC-EPs although the activation of VEGFR2/Akt or VEGFR2/p42/44MAPK pathway. Our study provided a new non-invasive source for endothelial progenitors, demonstrated critical roles of GREM1 in hUiPSC-EP and afforded a novel strategy to improve stem cell-based therapy for the ischaemic diseases.


Assuntos
Diferenciação Celular/genética , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linhagem Celular , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Imunofenotipagem , Modelos Biológicos , RNA Interferente Pequeno/genética
2.
Cell Commun Signal ; 18(1): 35, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127022

RESUMO

BACKGROUND: Inflammation and oxidative stress induced by oxidized low density lipoprotein are the main causes of vascular endothelial injury and atherosclerosis. Endothelial cells are important for the formation and repair of blood vessels. However, the detailed mechanism underlying the regulation of maturity and antioxidation of stem cell-derived endothelial like cells remains unclear. Besides, YY1 and TET2 play a key role on epigenetic modifications of proliferation and differentiation of stem cells. However, the regulatory mechanism of epigenetic modification induced by YY1 and TET2 on stem cells to iECICs is also not clear. AIM: Here, we want to investigate detailed mechanism underlying the regulation of maturity and antioxidation of stem cell-derived iECICs by by YY1 and TET2. METHODS: The qPCR, Western blot, immunohistochemical staining and flow cytometric analysis were used to analyze the expression level of each gene. Luciferase reporter assay was used to detect the binding sites between microRNA and target genes. The hMeDIP-sequence, ChIP-PCR and dot blot were used to detect the 5-hydroxymethylcytosine modification of genomic DNA. ATP, ROS, SOD assay were used to evaluate of oxidative stress in cells. The iECICs transplantation group The ApoE-/- mice were intravenous injected of iECICs to evaluation of therapeutic effect in vivo. RESULTS: Our studies have found that as the differentiation of human amniotic epithelial cells (HuAECs) is directed towards iECICs in vitro, the expression levels of vascular endothelial cell markers and miR-544 increase significantly and the expression level of YinYang 1 (YY1) decreases significantly. The luciferase reporter assay suggests that Yy1 is one of the targets of miR-544. Hydroxymethylated DNA immunoprecipitation sequencing showed that compared with HuAECs, iECICs had 174 protein-coding DNA sequences with extensive hydroxymethylation modifications. Overexpression of miR-544 inhibits the activity of the YY1/PRC2 complex and promotes the transcription and expression of the ten-eleven translocation 2 (TET2) gene, thereby activating the key factors of the serotonergic synapse pathway, CACNA1F, and CYP2D6. In addition, it promotes ability of maturity, antioxidation and vascular formation in vitro. Meanwhile, transplantation for miR-544-iECICs can significantly relieve oxidative stress injury on ApoE-/- atherosclerotic mice in vivo. CONCLUSIONS: miR-544 regulates the maturity and antioxidation of iECICs derived from HuAECs by regulating the YY1/TET2/serotonergic synapse signalling axis. Video abstract.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Epiteliais , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Dioxigenases , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout para ApoE , Estresse Oxidativo , Gravidez , Células-Tronco
3.
J Cell Mol Med ; 23(4): 0, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30734494

RESUMO

Tumour necrotic factor receptor-2 (TNFR2) has been to be cardiac-protective and is expressed in cardiac progenitor cells. Our goal is to define the mechanism for TNFR2-mediated cardiac stem cell activation and differentiation. By employing a protocol of in vitro cardiac stem cell (CSC) differentiation from human inducible pluripotent stem cell (hiPSC), we show that expression of TNFR2 precedes expression of CSC markers followed by expression of mature cardiomyocyte proteins. Activation of TNFR2 by a specific agonist promotes whereas inhibition of TNFR2 by neutralizing antibody diminishes hiPSC-based CSC differentiation. Interestingly, pluripotent cell factor RNA-binding protein Lin28 enhances TNFR2 protein expression in early CSC activation by directly binding to a conserved Lin28-motif within the 3'UTR of Tnfr2 mRNA. Furthermore, inhibition of Lin28 blunts TNFR2 expression and TNFR2-dependent CSC activation and differentiation. Our study demonstrates a critical role of Lin28-TNFR2 axis in CSC activation and survival, providing a novel strategy to enhance stem cell-based therapy for the ischaemic heart diseases.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Proteínas de Ligação a RNA/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Células Cultivadas , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais
4.
J Cell Biochem ; 120(8): 14165-14174, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30963639

RESUMO

Prostate cancer (PCa) is the third most common reason of cancer-related deaths in men. Accumulating evidence has shown that dysregulation of long noncoding RNAs (lncRNAs) is closely related to cancer initiation and development. Although large numbers of lncRNAs have been discovered, knowledge regarding their function and physiological/pathological significance remains limited. In this study, we aimed to reveal functional lncRNAs and identify prognosis-related RNAs in PCa by analyzing data from The Cancer Genome Atlas (TCGA). To achieve this, an lncRNA-mRNA coexpression network was constructed by weighted correlation network analysis. Additionally, a subnetwork was extracted from this weighted correlation network, and seven lncRNAs were identified as core nodes. Further Kaplan-Meier survival analysis showed that three lncRNAs (LINC00683, LINC00857, and FENDRR) were significantly downregulated in PCa samples, and there was a strong positive correlation with patient survival. Importantly, LINC00683 has not been fully reported as related with PCa. Additionally, gene set enrichment analysis indicated that LINC00683 might be involved in cancer-related pathways such as the Wnt pathway. Based on the findings of this study, lncRNA LINC00683 is likely to provide a new diagnostic biomarker and therapeutic target for future PCa treatments.


Assuntos
Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Humanos , Masculino , Prognóstico , Mapas de Interação de Proteínas/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Regulação para Cima/genética , Via de Sinalização Wnt/genética
5.
Cell Physiol Biochem ; 47(5): 1785-1799, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29953975

RESUMO

Backgroud/Aims: Mesenchymal stromal cells (MSCs) are a major component of the tumor microenvironment (TME). Several studies focusing on tumor-derived MSCs have demonstrated that they exhibit a strong ability to promote the tumor epithelial-mesenchymal transition (EMT). However, the factors mediating these effects are poorly understood. METHODS: Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunohistochemistry assays were used to detect the expression of Gremlin1 (GREM1) in human esophageal squamous cell carcinoma (ESCC) tissues. ShRNA silencing, flow cytometry, cell counting kit (CCK8) assay, invasion assay, western blot were used to detect the effect of GREM1 in ECa109, TE-1 cell lines and xenograft tumor models. RESULTS: In the current study, we found that the GREM1 was overexpressed in human ESCC tissues. The conditioned medium from mesenchymal stromal cells (MSCs-CM) enhanced the malignancy of xenograft esophageal tumors in vivo, as well as the cell proliferation, viability and invasion of the esophageal carcinoma cell lines ECa109 and TE-1 in vitro. Furthermore, the shRNA silencing of GREM1 in MSCs (shGREM1-MSCs) reversed the increased malignancy of the esophageal tumor in vivo, while the conditioned medium from shGREM1-MSCs (shGREM1-MSCs-CM) affected the cell cycle and cell invasion in vitro. These processes were accompanied by the EMT in the ECa109 and TE-1 cell lines with an alteration in the expression levels of mesenchymal and epithelial markers. Furthermore, the TGF-ß/BMP (transforming growth factor-beta/bone morphogenetic protein) signaling pathway participated in the shGREM1-MSCs-CM-induced anti-tumor effect on enhanced esophageal malignancy induced by MSCs-CM treatment. CONCLUSIONS: Taken together, our study suggested that GREM1 delivered by MSCs promoted EMT in ESCC in vitro and in vivo, which is partly through TGF-ß/BMP signaling pathway. The results provide experimental evidence to a potential therapeutic target in the treatment of esophageal cancer.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias Esofágicas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas de Neoplasias/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Neoplasias Esofágicas/patologia , Humanos , Metaloproteinases da Matriz Secretadas/metabolismo , Células-Tronco Mesenquimais/patologia , Fator de Crescimento Transformador beta/metabolismo
6.
J Cell Physiol ; 232(5): 996-1007, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27579673

RESUMO

Mesenchymal stem cells (MSCs) are a promising cell resource for the treatment of ischemic diseases, partially through paracrine effects. One of the major obstacles of MSC treatment is the poor survival rate and low efficiency of transplanted stem cells due to ischemic or inflammatory environments. Gremlin1 (GREM1), a regulator of growth, differentiation and development, has been identified as a novel proangiogenic factor. However, the role and mechanism of GREM1 in MSCs remains unclear. Therefore, we assessed the putative beneficial effects of GREM1 on MSC-based therapy for hindlimb ischemia. The lentiviral vector, EF1a-GREM1, was constructed using the Multisite Gateway System and used to transduce MSCs. In vitro studies demonstrated increased survival of GREM1-MSCs exposed to H2 O2 , which is consistent with the activation of caspase-3. Conditional medium from GREM1-MSCs (GREM1-MSC-CM) increased the anti-apoptotic effects of human umbilical vein endothelial cells (HUVECs), and this effect was attenuated by treatment with the PI3K/Akt pathway inhibitor LY294002. MSCs modified with GREM1 could significantly increase blood perfusion of the ischemic hindlimb in vivo in a mouse model, which was correlated to improved MSC survival. This study demonstrates that overexpression of GREM1 in MSCs have greater therapeutic effects against ischemia compared with wild-type MSCs by enhancing the survival of MSCs and ECs, which may provide new tools for studies investigating the treatment of ischemic diseases. J. Cell. Physiol. 232: 996-1007, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isquemia/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Apoptose , Arteríolas/patologia , Capilares/patologia , Sobrevivência Celular , Embrião de Galinha , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Isquemia/patologia , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neovascularização Fisiológica , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fluxo Sanguíneo Regional , Transdução de Sinais , Doadores de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
J Cell Mol Med ; 16(2): 296-305, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21418517

RESUMO

The mechanisms of progesterone on endothelial cell motility are poorly investigated. Previously we showed that progesterone stimulated endothelial cell migration via the activation of actin-binding protein moesin, leading to actin cytoskeleton remodelling and the formation of cell membrane structures required for cell movement. In this study, we investigated the effects of progesterone on the formation of focal adhesion complexes, which provide anchoring sites for cell movement. In cultured human umbilical endothelial cells, progesterone enhanced focal adhesion kinase (FAK) phosphorylation at Tyr(397) in a dose- and time-dependent manner. Several signalling inhibitors interfered with progesterone-induced FAK activation, including progesterone receptor (PR) antagonist ORG 31710, specific c-Src kinase inhibitor PP2, phosphatidylinosital-3 kinase (PI3K) inhibitor wortmannin as well as ρ-associated kinase (ROCK-2) inhibitor Y27632. It suggested that PR, c-Src, PI3K and ROCK-2 are implicated in this action. In line with this, we found that progesterone rapidly promoted c-Src/PI3K/Akt activity, which activated the small GTPase RhoA/ρ-associated kinase (ROCK-2) complex, resulting in FAK phosphorylation. In the presence of progesterone, endothelial cells displayed enhanced horizontal migration, which was reversed by small interfering RNAs abrogating FAK expression. In conclusion, progesterone promotes endothelial cell movement via the rapid regulation of FAK. These findings provide new information on the biological actions of progesterone on human endothelial cells that are relevant for vascular function.


Assuntos
Células Endoteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Progesterona/metabolismo , Amidas/farmacologia , Androstadienos/farmacologia , Proteína Tirosina Quinase CSK , Movimento Celular , Células Cultivadas , Estrenos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/genética , Adesões Focais/metabolismo , Furanos/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptores de Progesterona/antagonistas & inibidores , Transdução de Sinais , Wortmanina , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Quinases da Família src
8.
Mol Biol Rep ; 39(8): 8177-85, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22570111

RESUMO

Metabolic syndrome (MBS), a cluster of metabolic abnormalities and visceral fat accumulation, increases cardiovascular risks in postmenopausal women. In addition to visceral fat, perivascular adipose tissue has been recently found to play an important role in vascular pathophysiology. Hence, the present study investigates the effects of estrogen on both intra-abdominal fat (visceral fat) and periaortic fat (perivascular fat) accumulation as well as hypoxia in ovariectomized female rats. Female rats were divided into sham operation, ovariectomy and ovariectomy with 17ß-estradiol supplementation groups. Twelve weeks later, we found that estrogen improved MBS via reducing body weight gain, the weight of periaortic and intra-abdominal fat, hepatic triglyceride, and total serum cholesterol levels. Estrogen also increased insulin sensitivity through restoring glucose and serum leptin levels. For periaortic fat, western blot showed estrogen inhibited hypoxia by reducing the levels of VEGF and HIF-1α, which is consistent with the results from immunohistochemical staining. The correlation analysis indicated that perivascular fat had a positive correlation with body weight, intra-abdominal fat or serum total cholesterol, but a negative correlation with insulin sensitivity index. For intra-abdominal fat, real-time fluorescent RT-PCR showed estrogen improved fat dysfunction via reducing the levels of relative leptin, MCP-1 but increasing adiponectin mRNA. Estrogen reduced the levels of VEGF and HIF-1α to inhibit hypoxia but restored the levels of PPARγ and Srebp-1c, which are important for lipid capacity function of intra-abdominal fat. These results demonstrated estrogen improved MBS through down-regulating VEGF and HIF-1α to inhibit hypoxia of periaortic and intra-abdominal fat in ovariectomized female rats.


Assuntos
Estrogênios/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Gordura Intra-Abdominal/metabolismo , Síndrome Metabólica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adiponectina/genética , Animais , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Quimiocina CCL2/genética , Colesterol/sangue , Regulação para Baixo/efeitos dos fármacos , Feminino , Insulina/sangue , Resistência à Insulina , Gordura Intra-Abdominal/efeitos dos fármacos , Leptina/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Síndrome Metabólica/genética , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , PPAR gama/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismo , Útero/efeitos dos fármacos , Útero/metabolismo
9.
Front Genet ; 13: 831162, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35559023

RESUMO

Accumulating evidence indicates that the N6-methyladenosine (m6A) modification plays a critical role in human cancers. Given the current understanding of m6A modification, this process is believed to be dynamically regulated by m6A regulators. Although the discovery of m6A regulators has greatly enhanced our understanding of the mechanism underlying m6A modification in cancers, the function and role of m6A in the context of prostate cancer (PCa) remain unclear. Here, we aimed to establish a comprehensive diagnostic scoring model that can act as a complement to prostate-specific antigen (PSA) screening. To achieve this, we first drew the landscape of m6A regulators and constructed a LASSO-Cox model using three risk genes (METTL14, HNRNP2AB1, and YTHDF2). Particularly, METTL14 expression was found to be significantly related to overall survival, tumor T stage, relapse rate, and tumor microenvironment of PCa patients, showing that it has important prognostic value. Furthermore, for the sake of improving the predictive ability, we presented a comprehensive diagnostic scoring model based on a novel 6-gene panel by combining with genes found in our previous study, and its application potential was further validated by the whole TCGA and ICGC cohorts. Our study provides additional clues and insights regarding the treatment and diagnosis of PCa patients.

10.
Stem Cell Res ; 58: 102626, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34942479

RESUMO

Hypertrophic cardiomyopathy (HCM) is characterized by impaired energy metabolism irrespective of the degree of hypertrophy. Here, we reprogrammed peripheral blood mononuclear cells (PBMCs) isolated from peripheral blood of a patient who suffered from hypertrophic cardiomyopathy, into pluripotent stem cells (iPSC) (SYSUi005-A) using Sendai-virus. The established hiPSC line displayed a normal 46XY karyotype, showing strong expression of pluripotency markers and differentiation potential. This cell line may represent a valuable tool for investigating the pathogenesis and therapeutic strategies of HCM.


Assuntos
Cardiomiopatia Hipertrófica , Células-Tronco Pluripotentes Induzidas , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Diferenciação Celular , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Mutação
11.
ACS Appl Mater Interfaces ; 14(32): 36289-36303, 2022 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-35920579

RESUMO

Although stem cell-derived exosomes have been recognized as new candidates for cell-free treatment in myocardial infarction (MI), the challenge to improve the exosome retention in ischemic tissue remains. Our previous research indicated that islet-1(ISL1) overexpression enhances the paracrine function of mesenchymal stem cells (MSCs) and promotes angiogenesis in a model of MI. In this study, genetically engineered ISL1-MSC-derived exosomes (ISL1-MSCs-Exo) were collected, and the contents were analyzed by exosomal RNA sequencing. Next, we investigated if ISL1-MSCs-Exo could exert therapeutic effects and their incorporation into a new angiogenin-1 hydrogel (Ang-1 gel) could boost the retention of exosomes and further enhance their protective effects. Our results demonstrated that ISL1-MSCs-Exo could play a therapeutic role in vitro and in vivo, which might be due to changed exosomal contents. Ang-1 gel increased the retention and enhanced the anti-apoptosis, proliferation, and angiogenic capacity of ISL1-MSCs-Exo in endothelial cells. Echocardiography revealed that Ang-1 gel significantly augment the therapeutic effects of ISL1-MSCs-Exo for MI. The main mechanism might result from increased retention of ISL1-MSCs-Exo, herein enhanced pro-angiogenetic effects in an ischemic heart. Taken together, our findings indicated that ISL1-MSCs-Exo had endothelium-protective and pro-angiogenic abilities alone and Ang-1 gel could notably retain ISL1-MSCs-Exo at ischemic sites, which improved the survival and angiogenesis of endothelial cells and accelerated the recovery of MI. These results not only shed light on the therapeutic mechanism of ISL1-MSCs-Exo incorporated with Ang-1 gel but also offer a promising therapeutic option for ischemic disease.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Infarto do Miocárdio , Células Endoteliais/metabolismo , Exossomos/metabolismo , Humanos , Hidrogéis/metabolismo , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Neovascularização Patológica/metabolismo , Ribonuclease Pancreático
12.
Mol Biol Rep ; 38(8): 4885-92, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21170593

RESUMO

Our findings indicate that in ovariectomized female rats abdominal aortic constriction led to significant increases in left ventricular mass, myocyte diameter and heart weight/body weight (HW/BW) value, and decreases in interventricular septal thickness at diastole (IVSd), left ventricular percent fractional shortening (FS) and ejection fraction (EF). These pathophysiological alterations were largely reversed by administration with 17ß-estradiol for eight weeks. Furthermore, the enhanced expression of extracellular signal-regulated kinases 1/2 and decreased expression of caveolin-3 were found in left ventricle of AAC group. 17ß-estradiol (E(2)) administration increased the expression of caveolin-3 and reduced the level of ERK phosphorylation in these pressure-overloaded rats. Moreover, in cultured neonatal rat cardiomyocytes, E(2) inhibited the hypertrophic response to angiotensin II. This effect was reinforced by the addition of extracellular signal-regulated kinases 1/2 inhibitor PD98059, but was impaired when the cells were pretreated with caveolae disruptor, methyl-ß-cyclodextrin (M-ß-CD). In conclusion, our data indicate that estrogen attenuates the hypertrophic response induced by pressure overload through down-regulation of extracellular signal-regulated kinases 1/2 phosphorylation and up-regulation of caveolin-3 expression.


Assuntos
Caveolina 3/metabolismo , Estradiol/farmacologia , Miocárdio/metabolismo , Miocárdio/patologia , Ovariectomia , Pressão , Animais , Western Blotting , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Eletrocardiografia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Hipertrofia , Miocárdio/enzimologia , Ratos , Ratos Sprague-Dawley , beta-Ciclodextrinas/farmacologia
13.
Pharmacology ; 86(2): 117-24, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20689342

RESUMO

BACKGROUND: Genistein (GST) is a phytoestrogen that binds estrogen receptors (ER) to produce a protective cardiovascular effect. It also has been shown binding peroxisome proliferator-activated receptor-gamma (PPAR-gamma). METHODS: In the present study, we assessed the role of PPAR-gamma and ER in GST-mediated regulation of vascular tone in female rat aortas by in vitro tension measurement, immunohistochemistry, immunofluorescence, immunoprecipitation and Western blot analysis. RESULTS: In aortas pretreated with GW9662 (inhibitor of PPAR-gamma), ICI182780 (inhibitor of ER) and a combination of GW9662 and ICI182780, the magnitudes of GST-induced dilatation were attenuated. N(G)-nitro-L-arginine methyl ester had a similar effect. ER-beta and PPAR-gamma colocalized in all 3 layers of the aortas, while ER-alpha and PPAR-gamma colocalized only in the vascular endothelium and adventitia. In GST-treated whole-cell protein samples, we demonstrated coimmunoprecipitation of ER-beta (not ER-alpha) and PPAR-gamma. The expression of ER-beta and PPAR-gamma in nuclear protein from GST-treated samples increased, which was partially reversed by either GW9662 or ICI182780 and more efficiently reversed using a combination of GW9662 and ICI182780. CONCLUSION: Our findings suggest that GST can relax phenylephrine-induced vascular contraction in female rat aortas, which is mediated by PPAR-gamma and ER-beta.


Assuntos
Aorta Torácica/efeitos dos fármacos , Receptor alfa de Estrogênio/fisiologia , Genisteína/farmacologia , PPAR gama/fisiologia , Fitoestrógenos/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Aorta Torácica/citologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/fisiologia , Feminino , Imuno-Histoquímica , Imunoprecipitação , Técnicas In Vitro , Óxido Nítrico/antagonistas & inibidores , Especificidade de Órgãos , PPAR gama/antagonistas & inibidores , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
14.
J Altern Complement Med ; 25(4): 435-440, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30431316

RESUMO

OBJECTIVE: To explore the association between green tea consumption and coronary arterial disease (CAD) in the Chinese population of Guangzhou. Design, location, subjects: A retrospective study at the Sun Yat-sen Memorial Hospital in Guangzhou, China. Consecutive patients were enrolled between January 2013 and August 2014. A total of 539 patients were included. Two hundred sixty-seven of them are CAD patients and 272 of them are non-CAD patients. The CAD patients were diagnosed according to international diagnostic criteria. INTERVENTIONS: Using data from the questionnaires and clinical laboratories, we attempted to elucidate the association between green tea and CAD. OUTCOME MEASURES: Baseline characteristics of study population, CAD-related biomarkers, amount, frequency and duration of green tea consumption, and CAD risk analysis. RESULTS: The results showed that among males, those who drank green tea did not have a reduced risk of CAD (odds ratio; OR = 1.58, 95% CI: 0.96-2.59, p > 0.05). However, women in the study who drank green tea had a reduced risk of CAD (OR = 0.13, 95% CI: 0.07-0.23, p < 0.01). The females who consumed ≤1 cup/day green tea had lower CAD risk (OR = 0.12, 95% CI: 0.07-0.23, p < 0.01). The frequency of 3-5 days/week (OR = 0.14, 95% CI: 0.07-0.29, p < 0.01) and >5 days/week (OR = 0.24, 95% CI: 0.08-0.69, p < 0.01) were both beneficial in preventing CAD. Those who had been drinking green tea for 0-10 years (OR = 0.11, 95% CI: 0.04-0.30), 10-20 years (OR = 0.22, 95% CI: 0.11-0.46), or >20 years (OR = 0.37, 95% CI: 0.12-0.96) had a reduced risk of CAD. CONCLUSIONS: Through the analysis of green tea consumption and CAD-related biomarkers, we concluded that a small amount of high-frequency green tea consumption was associated with a reduced risk of CAD in female populations in Guangzhou, China, and the association might be partly due to altered CAD-related biomarkers.


Assuntos
Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/epidemiologia , Fitoterapia/estatística & dados numéricos , Chá , Idoso , China/epidemiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
15.
Nutr Res Pract ; 13(5): 393-398, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31583058

RESUMO

BACKGROUND/OBJECTIVES: The association between tea consumption and risk of coronary heart disease (CHD) remains controversial. This study aimed to determine whether tea consumption has an effect on CHD risk in Chinese adults. SUBJECTS/METHODS: In this hospital-based case-control study, 267 cases of CHD and 235 non-CHD controls were enrolled. Blood samples from all cases were examined. Cardiac function indices (left ventricular ejection fraction, left ventricular end-diastolic dimension, lactate dehydrogenase, and creatine kinase of the muscle or brain type), blood lipid index (high-density lipoprotein cholesterol), and blood coagulation function indices (fibrinogen and activated partial thromboplastin time) were recorded. Tea consumption of study participants was assessed by a specifically designed questionnaire. The baseline characteristics of the study populations were recorded, and CHD-related biomarkers were detected. Differences in baseline characteristics of the study participants were examined using t-tests for continuous variables and chi-squared tests for categorical variables. Unconditional logistic regression was used to measure the association between tea and CHD. RESULTS: There were significant differences in cardiac function indices, blood lipid index, and blood coagulation indices between CHD cases and controls (P < 0.05). We found tea consumption reduced CHD risk in female participants (adjusted odds ratio (OR) = 0.484, 95% CI: 0.242-0.968, P = 0.0403). Regarding the type of tea consumed, the risk of CHD was reduced in women who drank partially fermented tea (adjusted OR = 0.210, 95% CI: 0.084-0.522, P = 0.0008). Analytic results for the amount of tea consumed per unit time showed CHD risk was reduced in women who consumed 1-2 cups of tea per day (adjusted OR = 0.291, 95% CI: 0.131-0.643, P = 0.0023). A tea-drinking frequency of > 6 days/week was beneficial for CHD prevention (adjusted OR = 0.183, 95% CI: 0.049-0.679, P = 0.0112). When analyzed according to the duration of tea consumption, the risk of CHD was reduced in participants who had been drinking tea for 10-20 years (adjusted OR = 0.360, 95% CI: 0.137-0.946, P = 0.0382). CONCLUSIONS: Tea consumption is associated with a reduced risk of CHD in female but not male populations in Guangzhou.

16.
Int J Cardiol Heart Vasc ; 23: 100317, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31321282

RESUMO

BACKGROUND: Controversy still exists that whether clopidogrel should add proton pump inhibitors (PPIs) in patients with coronary heart disease after percutaneous coronary intervention (PCI). The aim of this study was to evaluate the efficacy and safety of clopidogrel added proton pump inhibitors (PPIs) vs. clopidogrel for the treatment of patients with coronary heart disease after percutaneous coronary intervention (PCI). METHODS AND RESULTS: We systematically searched PubMed, EMBASE, Web of Science, the Chinese Biomedical Medical Literature database, and the Cochrane Library for all clinical trials that were published on this topic through October 2018. We specifically selected the clinical trials that evaluated the efficacy and safety of clopidogrel added proton pump inhibitors vs. clopidogrel in the treatment of patients with coronary heart disease after PCI. RevMan 5.0 software was used for quantitative data analyses.15 randomized controlled trials including 50,366 patients were included. The meta-analysis results showed that compared with the clopidogrel added PPI group, the non-PPI group had significantly less risk of MACE[RR = 0.82,95%CI:0.77-0.88], myocardial infarction recurrence[RR = 0.72,95%CI:0.57-0.90], stent thrombosis[RR = 0.71,95%CI:0.56-0.92], Target vessel revascularization (TVR)[RR = 0.77,95%CI:0.63-0.93] and stroke [RR = 0.72,95%CI:0.67-0.76]. The risks of all cause death [RR = 1.14,95%CI:0.85-1.51], cardiovascular death [RR = 1.14, 95% CI: 0.85-1.52], bleedings events [RR = 1.60,95%CI:0.53-4.81] were similar in the two groups. CONCLUSIONS: The patients in the non-PPI group were observed to be associated with less risk of MACE, myocardial infarction recurrence, stent thrombosis, target vessel revascularization (TVR) and stroke. And the two groups had similar all cause death, cardiovascular death, bleedings events.

17.
Stem Cell Res Ther ; 9(1): 51, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29482621

RESUMO

BACKGROUND: The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed as a marker for cardiovascular progenitor cells. This study investigated whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) improves myocardial infarction (MI) treatment outcomes. METHODS: The lentiviral vector containing the human elongation factor 1α promoter, which drives the expression of ISL1 (EF1α-ISL1), was constructed using the Multisite Gateway System and used to transduce hMSCs. Flow cytometry, immunofluorescence, Western blotting, TUNEL assay, and RNA sequencing were performed to evaluate the function of ISL1-overexpressing hMSCs (ISL1-hMSCs). RESULTS: The in vivo results showed that transplantation of ISL1-hMSCs improved cardiac function in a rat model of MI. Left ventricle ejection fraction and fractional shortening were greater in post-MI hearts after 4 weeks of treatment with ISL1-hMSCs compared with control hMSCs or phosphate-buffered saline. We also found that ISL1 overexpression increased angiogenesis and decreased apoptosis and inflammation. The greater potential of ISL1-hMSCs may be attributable to an increased number of surviving cells after transplantation. Conditioned medium from ISL1-hMSCs decreased the apoptotic effect of H2O2 on the cardiomyocyte cell line H9c2. To clarify the molecular basis of this finding, we employed RNA sequencing to compare the apoptotic-related gene expression profiles of control hMSCs and ISL1-hMSCs. The results showed that insulin-like growth factor binding protein 3 (IGFBP3) was the only gene in ISL1-hMSCs with a RPKM value higher than 100 and that the difference fold-change between ISL1-hMSCs and control hMSCs was greater than 3, suggesting that IGFBP3 might play an important role in the anti-apoptosis effect of ISL1-hMSCs through paracrine effects. Furthermore, the expression of IGFBP3 in the conditioned medium from ISL1-hMSCs was almost fourfold greater than that in conditioned medium from control hMSCs. Moreover, the IGFBP3 neutralization antibody reversed the apoptotic effect of ISL1-hMSCs-CM. CONCLUSIONS: These results suggest that overexpression of ISL1 in hMSCs promotes cell survival in a model of MI and enhances their paracrine function to protect cardiomyocytes, which may be mediated through IGFBP3. ISL1 overexpression in hMSCs may represent a novel strategy for enhancing the effectiveness of stem cell therapy after MI.


Assuntos
Proteínas com Homeodomínio LIM/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/terapia , Fatores de Transcrição/genética , Animais , Células Cultivadas , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/metabolismo , Comunicação Parácrina , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
19.
Int J Cardiol ; 158(1): 46-53, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21255851

RESUMO

BACKGROUND: The mechanism that estrogen (E(2)) increases the number of endothelial progenitor cells (EPC) is largely unknown. Here we used E(2)-conjugated bovine serum albumin (E(2)-BSA, membrane impermeable) to investigate whether the membrane estrogen receptor (mER) and its related protein caveolin-1 (CAV-1) are involved in these processes. METHODS AND RESULTS: E(2)-BSA promoted [(3)H]-thymidine incorporation of EPC through increasing CAV-1 expression via mER (ERα, but not ERß or GPR30). Both cholesterol depletion and CAV-1 knockdown with use of CAV-1 siRNA significantly attenuated E(2)-BSA-induced [(3)H]-thymidine incorporation. Western blot showed that E(2)-BSA increased membrane CAV-1 protein expression 12h after treatment, whereas mRNA levels of CAV-1 were augmented until 24h after E(2)-BSA treatment. Furthermore, pre-incubated EPC with ICI 182780 (a specific ER antagonist), LY 294002 (a selective PI(3)K inhibitor) or PD 98059 (a specific ERK1/2 inhibitor) before E(2)-BSA inhibited the late-stage effect of E(2)-BSA (≥24 h) on up-regulation of CAV-1 mRNA and protein expression. Pulse chase results demonstrated that E(2)-BSA inhibited lysosome-mediated degradation of CAV-1 protein at the early stage (≤12 h), and then resulted in the increased CAV-1 protein. CONCLUSION: In the present work we demonstrated that E(2)-BSA promotes EPC proliferation through mER (ERα) in CAV-1-dependent manner: prolonging the stability of CAV-1 protein through quick inhibition of the lysosomal degradation pathway at the early stage (≤12 h) and up-regulating CAV-1 at transcription levels through PI(3)K/ERK1/2 signaling pathway at the late stage (≥24 h). These data indicated that a there is a novel mechanism of E(2)-BSA in the regulation of EPC proliferation through CAV-1.


Assuntos
Caveolina 1/fisiologia , Proliferação de Células , Células Endoteliais/citologia , Estradiol/fisiologia , Lisossomos/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Soroalbumina Bovina/fisiologia , Animais , Feminino , Ratos , Ratos Sprague-Dawley , Células-Tronco
20.
J Altern Complement Med ; 18(2): 143-52, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22339103

RESUMO

BACKGROUND: Individuals with prehypertension are at risk of hypertension and cardiovascular diseases, and yet efficient interventions are lagging behind. Studies indicate that heart rate variability-biofeedback (HRV-BF) increases HRV and baroreflex sensitivity (BRS) as well as reduces related pathological symptoms, suggesting potentially beneficial effects of HRV-BF on prehypertension, but little is known about these effects. In this study, these effects were investigated and their mechanisms were explored. OBJECTIVES: The effect of HRV-BF on prehypertension in young adults and its potential mechanism were explored. DESIGN: Forty-three (43) individuals with prehypertension were recruited and classified into three categories: HRV-BF group, slow abdominal breathing group, and control group. All groups were assessed with measurements of noninvasive blood pressure (BP), BRS, respiration, and galvanic skin response (GSR) at pre-intervention, in the entire process of each session, at postintervention, as well as at a 3-month follow-up. INTERVENTIONS: Subjects participated in a 10-session HRV-BF protocol or simple slow abdominal breathing protocol conducted over 5 weeks. A 3-month follow-up was also performed on these individuals. RESULTS: The incidence of prehypertension was as high as 14.5% in young college students. Individuals with prehypertension were lower in BRS (7.5±5.2 ms/mm Hg) and HRV (log10-transformed of the standard deviation of normal-to-normal beats [SDNN]=1.62±0.13 ms, lgTotal power of spectral density in the range of frequencies between 0 and 0.4Hz (TP)=8.02±0.55 ms2) than those with normal blood pressure (BRS=18.4±7.4 ms/mm Hg, lgSDNN=1.79±0.10 ms, lgTP=8.68±0.85 ms2). HRV-BF reduced blood pressure (from 131.7±8.7/79.3±4.7 mm Hg to 118.9±7.3 mm Hg/71.9±4.9 mm Hg, p<0.01), increased BRS (from 7.0±5.9 ms/mm Hg to 15.8±5.3 ms/mm Hg, p<0.01) and increased HRV (lgSDNN from 1.61±0.11 to 1.75±0.05 ms, and lgTP from 8.07±0.54 to 9.08±0.41 ms2, p<0.01). These effects were more obvious than those of the slow-breathing group, and remained for at least 3 months. HRV-BF also significantly increased vagus-associated HRV indices and decreased GSR (indices of sympathetic tone). CONCLUSIONS: These effects suggest that HRV-BF, a novel behavioral neurocardiac intervention, could enhance BRS, improve the cardiac autonomic tone, and facilitate BP adjustment for individuals with prehypertension.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Barorreflexo/fisiologia , Biorretroalimentação Psicológica , Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Pré-Hipertensão/terapia , Adulto , Exercícios Respiratórios , Feminino , Seguimentos , Resposta Galvânica da Pele , Humanos , Masculino , Pré-Hipertensão/fisiopatologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA