Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
2.
Adipocyte ; 10(1): 38-47, 2021 12.
Article in English | MEDLINE | ID: mdl-33371766

ABSTRACT

Human adipose-derived stem cells (hADSCs) are adult mesenchymal cells that have attracted the interest of clinical scientists and surgeons due to their large number of advantages including ease of access and expansion, abundance in cell culture, high proliferative rates, and lower senescence. SUMO/sentrin specific protease 1 (SENP1) is a critical protease that is required during the process of SUMOylation and deSUMOylation, which are dynamic mechanisms that influence cell cycle progression, cell proliferation, and apoptotic status. However, the contribution of SENP1 to these important cellular processes in hADSCs is largely uncharacterized and further studies in this area are required. Here, we show for the first time that after knock out SENP1 in hADSCs, their capacity to migrate and proliferate were inhibited, while apoptosis was enhanced. However, SENP1 did not significantly influence the morphology and MSC-related phenotypes of the hADSCs. These results highlight a role for SENP1 during hADSC growth, and its potential as a therapeutic target to improve the efficacy and safety of hADSCs in the clinic.


Subject(s)
Cysteine Endopeptidases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , China , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/physiology , Female , Humans , Male , Mesenchymal Stem Cells/physiology , Peptide Hydrolases/metabolism , Signal Transduction , Sumoylation
3.
Biochem Biophys Res Commun ; 527(3): 791-798, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32430183

ABSTRACT

Transplantation of endothelial progenitor cells (EPCs) has high therapeutic potential for ischemia-related ailments like heart attacks and claudication. Due to limited EPC sources, direct reprogramming is a fast-developing way to convert human-induced pluripotent stem cells (hiPSCs) into EPCs fit for transplantation. However, the procedural efficacy was affected by multiple factors, including epigenetic modifications. Recent studies have shown that m7G methylation mediated by Methyltransferase like 1 (METTL1) is required for mouse embryonic stem cells (mESCs) to differentiate normally. Yet, its contributions to EPC differentiation still require elucidation. Here, using immunofluorescence microscopy and Fluorescence-activated Cell Sorting (FACS), we found that the typical EPC markers were significantly increased in METTL1 knockdown (METTL1-KD) hiPSCs-derived EPCs compared to those of control types. In addition, we found that METTL1 knockdown activates the MAPK/ERK signaling pathway during EPCs differentiation from hiPSCs. Furthermore, functional properties of METTL1-KD EPCs were significantly raised above those of control hiPSCs-derived EPCs. Moreover, we proved that METTL1-KD hiPSCs-derived EPCs significantly accelerate vascular smooth muscle cell proliferation and 'phenotype switching' through a co-culture system. To sum up, our results demonstrate that METTL1-KD significantly promotes the differentiation of EPCs along with their in vitro functions, and this effect may be achieved through activation of the MAPK/ERK signaling pathway. This enhances current knowledge of EPC generation from hiPSCs and presents a new therapeutic target of vascular diseases.


Subject(s)
Endothelial Progenitor Cells/cytology , Induced Pluripotent Stem Cells/cytology , MAP Kinase Signaling System , Methyltransferases/metabolism , Cell Differentiation , Cell Line , Cell Proliferation , Endothelial Progenitor Cells/metabolism , Gene Knockdown Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Methyltransferases/genetics
4.
Braz J Med Biol Res ; 53(3): e9201, 2020.
Article in English | MEDLINE | ID: mdl-32130294

ABSTRACT

Methylophiopogonanone A (MO-A), a homoisoflavonoid extracted from Ophiopogon japonicus, has been shown to attenuate myocardial apoptosis and improve cerebral ischemia/reperfusion injury. However, the hypolipidemic effects remain unknown. This study was performed to investigate a potential hypolipidemic effect of MO-A in hyperlipidemia rats, as well as its underlying mechanism of action. A rat model of hyperlipidemia was induced by a high-fat diet (HFD). Animals were randomly divided into three groups (n=8/group): normal control group (NC), HFD group, and HFD+MO-A (10 mg·kg-1·d-1) treatment group. The effects of MO-A on serum lipids, body weight, activity of lipoprotein metabolism enzyme, and gene expression of lipid metabolism were evaluated in HFD-induced rats. In HFD-induced rats, pretreatment with MO-A decreased the body weight gain and reduced serum and hepatic lipid levels. In addition, pretreatment with MO-A improved the activities of lipoprotein lipase and hepatic lipase in serum and liver, down-regulated mRNA expression of acetyl CoA carboxylase and sterol regulatory element-binding protein 1c, and up-regulated mRNA expression of low-density lipoprotein receptor and peroxisome proliferator-activated receptor α in the liver. Our results indicated that MO-A showed strong ability to ameliorate the hyperlipidemia in HFD-induced rats. MO-A might be a potential candidate for prevention of overweight and dyslipidemia induced by HFD.


Subject(s)
Benzodioxoles/pharmacology , Diet, High-Fat , Hyperlipidemias/prevention & control , Isoflavones/pharmacology , Lipid Metabolism , Ophiopogon/chemistry , Animals , Benzodioxoles/isolation & purification , Blotting, Western , Disease Models, Animal , Feces/chemistry , Hyperlipidemias/metabolism , Isoflavones/isolation & purification , Lipids/analysis , Male , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
5.
Biochem Biophys Res Commun ; 522(4): 838-844, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31801667

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the authors. The authors found that the experiment to show that HG decreased CD31 (Figure 1 E) and increased Vimentin (Figure 1 F) mRNA level could not be repeated. Moreover, they found that HG had no effect on Ch25h protein level and cannot repeat the experiments that HG decreased Ch25h mRNA level (Figure 1 G). In summary, they cannot conclude that HG suppressed EndoMT through KLF4/Ch25h, which made great weakening to the protection of KLF4/Ch25h axis in blood glucose dysfunction-induced endothelial dysfunction and diabetes. The authors wish to apologize for any inconvenience they may have caused.


Subject(s)
Endothelium/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Mesoderm/metabolism , Metformin/pharmacology , Steroid Hydroxylases/metabolism , Animals , Endothelium/drug effects , Epigenesis, Genetic/drug effects , Glucose/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Kruppel-Like Factor 4 , Liver X Receptors/metabolism , Male , Mesoderm/drug effects , Mice, Inbred C57BL , Signal Transduction/drug effects
6.
Braz. j. med. biol. res ; 53(3): e9201, 2020. tab, graf
Article in English | LILACS | ID: biblio-1089347

ABSTRACT

Methylophiopogonanone A (MO-A), a homoisoflavonoid extracted from Ophiopogon japonicus, has been shown to attenuate myocardial apoptosis and improve cerebral ischemia/reperfusion injury. However, the hypolipidemic effects remain unknown. This study was performed to investigate a potential hypolipidemic effect of MO-A in hyperlipidemia rats, as well as its underlying mechanism of action. A rat model of hyperlipidemia was induced by a high-fat diet (HFD). Animals were randomly divided into three groups (n=8/group): normal control group (NC), HFD group, and HFD+MO-A (10 mg·kg-1·d-1) treatment group. The effects of MO-A on serum lipids, body weight, activity of lipoprotein metabolism enzyme, and gene expression of lipid metabolism were evaluated in HFD-induced rats. In HFD-induced rats, pretreatment with MO-A decreased the body weight gain and reduced serum and hepatic lipid levels. In addition, pretreatment with MO-A improved the activities of lipoprotein lipase and hepatic lipase in serum and liver, down-regulated mRNA expression of acetyl CoA carboxylase and sterol regulatory element-binding protein 1c, and up-regulated mRNA expression of low-density lipoprotein receptor and peroxisome proliferator-activated receptor α in the liver. Our results indicated that MO-A showed strong ability to ameliorate the hyperlipidemia in HFD-induced rats. MO-A might be a potential candidate for prevention of overweight and dyslipidemia induced by HFD.


Subject(s)
Animals , Male , Rats , Ophiopogon/chemistry , Benzodioxoles/pharmacology , Lipid Metabolism , Diet, High-Fat , Hyperlipidemias/prevention & control , Isoflavones/pharmacology , Blotting, Western , Rats, Sprague-Dawley , Disease Models, Animal , Benzodioxoles/isolation & purification , Feces/chemistry , Real-Time Polymerase Chain Reaction , Hyperlipidemias/metabolism , Isoflavones/isolation & purification , Lipids/analysis
7.
Water Sci Technol ; 77(11-12): 2668-2676, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29944131

ABSTRACT

In the present study, copper tailings were treated at different temperatures (50-650 °C) and for various times (0.5-6 hours) and their phosphate adsorption capacity was investigated. The results showed that heating temperature significantly affected adsorption capacity. The highest capacity was observed in treatments at 310-350 °C. Heating time did not influence phosphate adsorption ability of copper tailings. Scanning electron microscopy, Barrett-Joyner-Halenda (BJH), and Fourier transform infrared spectroscopy (FTIR) were employed to characterize untreated copper tailings (raw CT) and copper tailings heated at 340 °C (CT340). The results showed that CT340 had a rougher surface, more and smaller pores, a larger surface area and higher FTIR transmittance than raw CT. These changes in texture might explain the increased phosphate adsorption of thermally modified copper tailings. Mathematical modeling showed that the Langmuir nonlinear model was the best fit to the current data. The maximum adsorption capacities of raw CT and CT340 were predicted as 2.08 mg/g and 14.25 mg/g at 298 K, pH 6.0, respectively.


Subject(s)
Copper/chemistry , Phosphates/chemistry , Water Pollutants, Chemical/chemistry , Adsorption , Hot Temperature , Microscopy, Electron, Scanning , Mining , Phosphates/isolation & purification , Spectroscopy, Fourier Transform Infrared/methods , Time Factors
8.
Biochem Biophys Res Commun ; 495(2): 1864-1870, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29225169

ABSTRACT

Orai1-dependent Ca2+ entry plays an essential role in inflammatory response through regulating T cell and macrophage activation and neutrophil infiltration. However, whether Orai1 Ca2+ entry contributes to endothelial activation, one of the early steps of vascular inflammation, remains elusive. In the present study, we observed that knockdown of Orai1 reduced, whereas overexpression of Orai1 potentiated, TNFα-induced expression of adhesion molecules such as ICAM-1 and VCAM-1 in HUVECs, and subsequently blocked adhesion of monocyte to HUVECs. In vivo, Orai1 downregulation attenuated TNFα-induced ICAM-1 and VCAM-1 expression in mouse aorta and the levels of pro-inflammatory cytokines in the serum. In addition, Orai1 knockdown also dramatically decreased the expression of pro-inflammatory cytokines and neutrophil infiltration in the lung after TNFα treatment, and thus protected lung tissue injury. Notably, among all isoforms of nuclear factor of activated T cells (NFATs), TNFα only triggered NFATc4 nuclear accumulation in HUVECs. Knockdown of Orai1 or inhibition of calcineurin prevented TNFα-induced NFATc4 nuclear translocation and reduced ICAM-1 and VCAM-1 expression in HUVECs. Overexpression of NFATc4 further enhanced ICAM-1 and VCAM-1 expression induced by TNFα. Our study demonstrates that Orai1-Ca2+-calcineurin-NFATc4 signaling is an essential inflammatory pathway required for TNFα-induced endothelial cell activation and vascular inflammation. Therefore, Orai1 may be a potential therapeutic target for treatment of inflammatory diseases.


Subject(s)
Aortitis/immunology , Calcineurin/immunology , Calcium/immunology , Cell Adhesion Molecules/immunology , Endothelium, Vascular/immunology , NFATC Transcription Factors/immunology , ORAI1 Protein/immunology , Animals , Aortitis/pathology , Cells, Cultured , Down-Regulation/immunology , Humans , Inflammation Mediators/immunology , Metabolic Networks and Pathways/immunology , Mice , Mice, Inbred C57BL
9.
J Cell Mol Med ; 21(5): 904-915, 2017 05.
Article in English | MEDLINE | ID: mdl-27878958

ABSTRACT

Increasing evidence supports that activation of store-operated Ca2+ entry (SOCE) is implicated in the chemoresistance of cancer cells subjected to chemotherapy. However, the molecular mechanisms underlying chemoresistance are not well understood. In this study, we aim to investigate whether 5-FU induces hepatocarcinoma cell death through regulating Ca2+ -dependent autophagy. [Ca2+ ]i was measured using fura2/AM dye. Protein expression was determined by Western blotting and immunohistochemistry. We found that 5-fluorouracil (5-FU) induced autophagic cell death in HepG2 hepatocarcinoma cells by inhibiting PI3K/AKT/mTOR pathway. Orai1 expression was obviously elevated in hepatocarcinoma tissues. 5-FU treatment decreased SOCE and Orai1 expressions, but had no effects on Stim1 and TRPC1 expressions. Knockdown of Orai1 or pharmacological inhibition of SOCE enhanced 5-FU-induced inhibition of PI3K/AKT/mTOR pathway and potentiated 5-FU-activated autophagic cell death. On the contrary, ectopic overexpression of Orai1 antagonizes 5-FU-induced autophagy and cell death. Our findings provide convincing evidence to show that Orai1 expression is increased in hepatocarcinoma tissues. 5-FU can induce autophagic cell death in HepG2 hepatocarcinoma cells through inhibition of SOCE via decreasing Orai1 expression. These findings suggest that Orai1 expression is a predictor of 5-FU sensitivity for hepatocarcinoma treatment and blockade of Orai1-mediated Ca2+ entry may be a promising strategy to sensitize hepatocarcinoma cells to 5-FU treatment.


Subject(s)
Calcium/metabolism , Carcinoma, Hepatocellular/metabolism , Fluorouracil/pharmacology , Liver Neoplasms/metabolism , ORAI1 Protein/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Carcinoma, Hepatocellular/pathology , Down-Regulation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
10.
J Am Soc Nephrol ; 27(10): 3063-3078, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26940090

ABSTRACT

Evidence supports an important role of Ca2+ release-activated Ca2+ channel protein 1 (Orai1)-mediated Ca2+ entry in the development of renal fibrosis, a common pathologic feature of CKDs that lead to ESRD, but the molecular mechanisms remain unclear. We determined the role of Orai1 calcium channel in renal fibrosis induced by high-fat diet and by unilateral ureteral obstruction. Mouse kidneys with fibrosis had higher levels of Orai1 protein expression than did kidneys without fibrosis. In vivo knockdown of Orai1 with adenovirus harboring Orai1-short hairpin RNA or inhibition of Orai1 with SKF96365 dramatically prevented renal fibrosis and significantly decreased protein expression of fibronectin, α­smooth muscle actin, and TGF­ß1 in the kidney cortex of ApoE-/- mice on a high-fat diet and in the obstructed kidneys of mice with unilateral ureteral obstruction. Compared with kidney biopsy specimens of patients with glomerular minimal change disease, those of patients with fibrotic nephropathy had higher expression levels of Orai1. In cultured human proximal tubule epithelial cells (HK2), knockdown of Orai1 Ca2+ channel with adenovirus-Orai1-short hairpin RNA markedly inhibited TGF-ß1-induced intracellular Ca2+ influx and phosphorylation of smad2/3. Knockdown or blockade of the Orai1 Ca2+ channel in HK2 cells also prevented epithelial-to-mesenchymal transition induced by TGF­ß1. In conclusion, blockade of the Orai1 Ca2+ channel prevented progression of renal fibrosis in mice, likely by suppressing smad2/3 phosphorylation and TGF-ß1-induced epithelial-to-mesenchymal transition. These results render the Orai1 Ca2+ channel a potential therapeutic target against renal fibrosis.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium/metabolism , Imidazoles/pharmacology , Kidney/pathology , ORAI1 Protein/antagonists & inhibitors , Animals , Cells, Cultured , Fibrosis/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Circ J ; 80(4): 1024-33, 2016.
Article in English | MEDLINE | ID: mdl-26911455

ABSTRACT

BACKGROUND: Previous work has demonstrated that the volume-regulated chloride channel is activated during foam cell formation, and inhibition of chloride movement prevents intracellular lipid accumulation. However, the mechanism explaining how chloride movement promotes foam cell formation is not clear. METHODS AND RESULTS: Foam cell formation was determined by Oil Red O staining. Western blotting and co-immunoprecipitation were used to examine protein expression and protein-protein interaction. [Cl(-)]iwas measured using 6-methoxy-N-ethylquinolinium iodide dye. The results showed that [Cl(-)]iwas decreased in monocytes/macrophages from patients with hypercholesterolemia and from apoE(-/-)mice fed with a high-fat diet. Lowering [Cl(-)]iupregulated scavenger receptor A (SR-A) expression, increased the binding and uptake of oxLDL, enhanced pro-inflammatory cytokine production and subsequently accelerated foam cell formation in macrophages from humans and mice. In addition, low Cl(-)solution stimulated the activation of JNK and p38 mitogen-activated protein kinases. Inhibition of JNK and p38 blocked Cl(-)reduced medium-induced SR-A expression and lipid accumulation. In contrast, reduction of [Cl(-)]ipromoted the interaction of SR-A with caveolin-1, thus facilitating caveolin-1-dependent SR-A endocytosis. Moreover, disruption of caveolae attenuated SR-A internalization, JNK and p38 activation, and ultimately prevented SR-A expression and foam cell formation stimulated by low Cl(-)medium. CONCLUSIONS: This data provide strong evidence that reduction of [Cl(-)]iis a critical contributor to intracellular lipid accumulation, suggesting that modulation of [Cl(-)]iis a novel avenue to prevent foam cell formation and atherosclerosis.


Subject(s)
Chlorides/metabolism , Foam Cells/metabolism , Hypercholesterolemia/metabolism , Animals , Apolipoproteins E/deficiency , Caveolin 1/genetics , Caveolin 1/metabolism , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Foam Cells/pathology , Hypercholesterolemia/chemically induced , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Arterioscler Thromb Vasc Biol ; 36(4): 618-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26916730

ABSTRACT

OBJECTIVE: To determine the role of orai1 store-operated Ca(2+) entry in foam cell formation and atherogenesis. APPROACH AND RESULTS: Acute administration of oxidized low-density lipoprotein (oxLDL) activates an orai1-dependent Ca(2+) entry in macrophages. Chelation of intracellular Ca(2+), inhibition of orai1 store-operated Ca(2+) entry, or knockdown of orai1 dramatically inhibited oxLDL-induced upregulation of scavenger receptor A, uptake of modified LDL, and foam cell formation. Orai1-dependent Ca(2+) entry induces scavenger receptor A expression and foam cell formation through activation of calcineurin but not calmodulin kinase II. Activation of nuclear factor of activated T cells is not involved in calcineurin signaling to foam cell formation. However, oxLDL dephosohorylates and activates apoptosis signal-regulating kinase 1 in macrophages. Orai1 knockdown prevents oxLDL-induced apoptosis signal-regulating kinase 1 activation. Knockdown of apoptosis signal-regulating kinase 1, or inhibition of its downstream effectors, JNK and p38 mitogen-activated protein kinase, reduces scavenger receptor A expression and foam cell formation. Notably, orai1 expression is increased in atherosclerotic plaques of apolipoprotein E(-/-) mice fed with high-cholesterol diet. Knockdown of orai1 with adenovirus harboring orai1 siRNA or inhibition of orai1 Ca(2+) entry with SKF96365 for 4 weeks dramatically inhibits atherosclerotic plaque development in high-cholesterol diet feeding apolipoprotein E(-/-) mice. In addition, inhibition of orai1 Ca(2+) entry prevents macrophage apoptosis in atherosclerotic plaque. Moreover, the expression of inflammatory genes in atherosclerotic lesions and the infiltration of myeloid cells into the aortic sinus plaques are decreased after blocking orai1 signaling. CONCLUSIONS: Orai1-dependent Ca(2+) entry promotes atherogenesis possibly by promoting foam cell formation and vascular inflammation, rendering orai1 Ca(2+) channel a potential therapeutic target against atherosclerosis.


Subject(s)
Anticholesteremic Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium/metabolism , Cholesterol/metabolism , Foam Cells/drug effects , Macrophages, Peritoneal/drug effects , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcineurin/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Chelating Agents/pharmacology , Calcium Signaling/drug effects , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Foam Cells/metabolism , Foam Cells/pathology , Humans , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipoproteins, LDL/pharmacology , MAP Kinase Kinase Kinase 5/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice, Knockout , ORAI1 Protein , Plaque, Atherosclerotic , RNA Interference , Scavenger Receptors, Class A/metabolism , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL