Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Med Sci Monit ; 25: 318-323, 2019 Jan 11.
Article in English | MEDLINE | ID: mdl-30631031

ABSTRACT

BACKGROUND Hypertension is a leading global disease, and myocardial fibrosis is an important adverse effect of hypertension, seriously threatening human health. The IL-6/STAT3 pathway and endothelin-1 (ET-1) were previously suggested to play a part in myocardial fibrosis. MATERIAL AND METHODS To investigate the role of Atorvastatin (Ato) in spontaneous hypertension, systolic blood pressure (SBP) and left ventricular mass index (LVMI) were measured, and Masson trichrome staining was performed. Furthermore, the relative protein levels of the IL-6/STAT3/ET-1 pathway were tested. RESULTS Ato prevented myocardial fibrosis in spontaneous hypertension rats, especially at the dosage of 50 mg/kg/d. The IL-6/STAT3 pathway was observed to be suppressed by Ato, and ET-1 level in myocardial tissues was also downregulated by Ato. The phosphorylation status of STAT3 was tested after Ato treatment, showing that Ato mainly stimulated the tyr-705 phosphorylation of STAT3. CONCLUSIONS Results of this study may help promote myocardial fibrosis therapy and provide insights into the IL-6/STAT3/ET-1-mediated mechanism in Ato-induced myocardial fibrosis inhibition.


Subject(s)
Atorvastatin/pharmacology , Hypertension/metabolism , Animals , Atorvastatin/metabolism , Blood Pressure , Cardiomyopathies/metabolism , Disease Models, Animal , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/prevention & control , Endothelin-1/metabolism , Fibrosis/pathology , Hypertension/complications , Interleukin-6/metabolism , Male , Myocardium/pathology , Phosphorylation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
2.
Cardiol J ; 26(4): 394-404, 2019.
Article in English | MEDLINE | ID: mdl-29718531

ABSTRACT

BACKGROUND: Recent studies have identified amphoterin-induced gene and open reading frame (AMIGO2). The role of AMIGO2 in tumour research is well-studied, but its role in ischemic heart diseases is seldom reported. In the present study, the role of AMIGO2 in myocardial infarction (MI) is under investigation for the first time. METHODS: For in vitro studies, cardiomyocytes (CMs) and endothelial cells (ECs) were isolated from both AMIGO2 knockout (KO) and WT mice. The apoptosis of CMs was tested after 48 h of ischemic stimulation. A proliferation test was implemented after 7 days of normoxic incubation and tube forma-tion on ECs. For in vivo studies, the MI model was built in mice hearts. Echocardiographic evaluation was performed at 3 days and 28 days post-MI, while the hemodynamics test was performed at 28 days post-MI. The histological results of the apoptosis, proliferation, angiogenesis and infarct zone assess-ments were determined using terminal deoxynucleotidyl transferase-mediated dUTP nick end-labelling (TUNEL) assay, Ki67 staining, a-SMA/CD31 immunostain and the Masson-Trichrome method, respectively. The expression changes of the Akt pathway and related proteins were confirmed using both quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. RESULTS: The present results demonstrated that AMIGO2 deficiency caused more CMs suffering apop-tosis, lower proliferation and less angiogenesis in vitro and in vivo. Weaker cardiac function and larger scar formation were detected in AMIGO2 KO mice, and increased expression of active-caspase-3 and decreased expression of PDK1, p-Akt, Bcl-2/Bax and VEGF occurred. CONCLUSIONS: Herein the findings indicate that AMIGO2 deficiency plays an attenuated cardio-pro-tective role in ischemic heart disease via inactivation of the PDK1/Pten/Akt pathway.


Subject(s)
Endothelial Cells/metabolism , Membrane Proteins/deficiency , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/deficiency , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Nerve Tissue Proteins/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Left
3.
Int J Mol Med ; 39(3): 569-578, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28204828

ABSTRACT

The transforming growth factor (TGF)-ß-inducible early gene-1 (TIEG1) plays a crucial role in modulating cell apoptosis and proliferation in a number of diseases, including pancreatic cancer, leukaemia and osteoporosis. However, the functional role of TIEG1 in the heart has not been fully defined. In this study, we first investigated the role of TIEG1 in ischaemic heart disease. For in vitro experiments, cardiomyocytes were isolated from both TIEG1 knockout (KO) and wile-type (WT) mice, and the apoptotic ratios were evaluated after a 48­h ischaemic insult. A cell proliferation assay was performed after 7 days of incubation under normoxic conditions. In addition, the angiogenic capacity of endothelial cells was determined by tube formation assay. For in vivo experiments, a model of myocardial infarction (MI) was established using both TIEG1 KO and WT mice. Echocardiography was performed at 3 and 28 days post-MI, whereas the haemodynamics test was performed 28 days post-MI. Histological analyses of apoptosis, proliferation, angiogenesis and infarct zone assessments were performed using terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling (TUNEL) staining, BrdU immunostaining, α-smooth muscle actin (α-SMA)/CD31 immunostaining and Masson's trichrome staining, respectively. Changes in the expression of related proteins caused by TIEG1 deficiency were confirmed using both reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. Our results demonstrated that the absence of TIEG1 prevented cardiomyocytes from undergoing apoptosis and promoted higher proliferation; it stimulated the proliferation of endothelial cells in vitro and in vivo. Improved cardiac function and less scar formation were observed in TIEG1 KO mice, and we also observed the altered expression of phosphatase and tensin homolog (Pten), Akt and Bcl-2/Bax, as well as vascular endothelial growth factor (VEGF). On the whole, our findings indicate that the absence of TIEG1 plays a cardioprotective role in ischaemic heart disease by promoting changes in Pten/Akt signalling.


Subject(s)
Early Growth Response Transcription Factors/deficiency , Kruppel-Like Transcription Factors/deficiency , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Apoptosis , Biomarkers , Cell Proliferation , Disease Models, Animal , Echocardiography , Endothelial Cells , Heart Function Tests , Mice , Mice, Knockout , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...