Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Food Sci ; 88(8): 3494-3506, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37326339

ABSTRACT

This study investigated the effects of extrusion on the physical properties of glutinous rice and addressed the challenges associated with its hardened texture and reduced taste in glutinous rice products by adding extruded glutinous rice to assess their anti-retrogradation effect compared with different improvers. Glutinous rice flour with different gelatinization degrees was obtained by changing the initial moisture content of glutinous rice grains before extrusion, and their physicochemical properties and the effect of adding them to rice products were analyzed. Results showed that with the increase in moisture content, the viscosity, water absorption index of extruded glutinous rice flour, and product viscosity increased, while the gelatinization degree, water solubility index, and product elasticity decreased, and the hardness of the rice products showed a trend of first decreasing and then increasing. Twenty percent moisture content of glutinous rice products showed the best properties mentioned above. The effects of adding different improvers on retrogradation degree, quality characteristics, microstructure, and moisture migration of glutinous rice products were analyzed by texture profile analysis, sensory evaluation, scanning electron microscopy, and low-field nuclear magnetic resonance. It was found that soybean polysaccharides, xanthan gum, and extruded glutinous rice flour had better anti-retrogradation effects, while colloid and soybean polysaccharides provided a tighter and more three-dimensional internal structure to the rice products. Our study showed that extruded glutinous rice flour had good anti-retrogradation properties and little effect on flavor and taste, but it would increase the roughness and viscosity of the products, which had advantages and disadvantages compared with other improvers.


Subject(s)
Oryza , Oryza/chemistry , Chemical Phenomena , Viscosity , Solubility , Water/chemistry , Flour/analysis
2.
Neurobiol Aging ; 45: 76-87, 2016 09.
Article in English | MEDLINE | ID: mdl-27459928

ABSTRACT

Reduction of protein phosphatase-2A (PP2A) activity is a common clinical feature of Alzheimer's disease and vascular dementia. In this study, we observed that chronic brain hypoperfusion induced by bilateral common carotid artery occlusion of rats led to PP2A inactivation based on the increase in tyrosine-307 phosphorylation and leucine-309 demethylation of PP2AC and the depression in PP2ABα. Knockdown of miR-195 using overexpression of its antisense molecule oligonucleotide (pre-AMO-miR-195) delivered by a lentivirus (lenti-pre-AMO-miR-195) increased tyrosine-307 phosphorylation and decreased both PP2ABα expression and leucine-309 methylation; these effects were prevented by the overexpression of miR-195 using lenti-pre-miR-195 and controlled by an increase in methylesterase (PME-1) and a decrease in leucine carboxyl methyltransferase-1. In vitro studies demonstrated that miR-195 regulated PME-1 expression by binding to the Ppme1 gene 3'-untranslated region (3'UTR) domain. Masking the miR-195 binding sites in the amyloid precursor protein (APP) and ß-site APP cleaving enzyme 1 genes prevented miR-195-induced leucine carboxyl methyltransferase-1 elevation. We concluded that the miR-195 downregulation in chronic brain hypoperfusion involved PP2A inactivity, which was mediated by the post-transcriptional regulation PME-1, APP, and ß-site APP cleaving enzyme 1 expression.


Subject(s)
Brain Ischemia/enzymology , Enzyme Activation/genetics , Gene Knockdown Techniques , MicroRNAs/genetics , Protein Phosphatase 2/metabolism , Animals , Carboxylic Ester Hydrolases/metabolism , Chronic Disease , Down-Regulation , Male , Protein O-Methyltransferase/metabolism , Rats, Sprague-Dawley
3.
J Biol Chem ; 290(32): 19641-52, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26100622

ABSTRACT

The JAK-STAT3 signaling pathway is one of the critical pathways regulating cell proliferation, differentiation, and apoptosis. Myocardin is regarded as a key mediator for the change of smooth muscle phenotypes. However, the relationship between STAT3 and myocardin in the vascular smooth muscle cell (VSMC) phenotypic switch has not been investigated. The goal of this study was to investigate the molecular mechanism by which STAT3 affects the myocardin-regulated VSMC phenotypic switch. Data presented in this study demonstrated that STAT3 was rapidly up-regulated after stimulation with VEGF. Inhibition of the STAT3 activation process impaired VSMC proliferation and enhanced the expression of VSMC contractile genes by increasing serum-response factor binding to the CArG-containing regions of VSMC-specific contractile genes. In contrast, the interaction between serum-response factor and its co-activator myocardin was reduced by overexpression of STAT3. In addition, treated VEGF inhibited the transcription activity of myocardin, and overexpression of STAT3 inhibited myocardin-induced up-regulation of VSMC contractile phenotype-specific genes. Although myocardin and STAT3 are negatively correlated, interestingly, both of them can enhance the expression of VEGF, suggesting a feedback loop to regulate the VSMC phenotypic switch. Taken together, these results indicate that the JAK-STAT3 signaling pathway plays a key role in controlling the phenotypic switch of VSMCs through the interactions between STAT3 and myocardin by various coordinated gene regulation pathways and feedback loops.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Phenotype , STAT3 Transcription Factor/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , Cell Differentiation , Cell Line , Cell Proliferation , Feedback, Physiological , Gene Expression Regulation , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Contraction/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/genetics , STAT3 Transcription Factor/genetics , Serum Response Factor/genetics , Signal Transduction , Trans-Activators/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
4.
IUBMB Life ; 67(3): 202-17, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25854163

ABSTRACT

Breast cancer is the leading cause of cancer death in women worldwide which is closely related to metastasis. But the exact molecular mechanism of metastasis is still not fully understood. We now report that both MRTF-A and STAT3 play important roles in migration of MDA-MB-231 breast cancer cells. Moreover, MRTF-A and STAT3 synergistically increased MDA-MB-231 cell migration by promoting the expression of migration markers urokinase-type plasminogen activator (uPA) and osteopontin (OPN) and inhibiting the expression of breast cancer metastasis suppressor 1 (BRMS1). Luciferase reporter assays demonstrated that MRTF-A and STAT3 do not affect transcription of the BRMS1 promoter. Instead, we identified a newly molecular mechanism by which MRTF-A and STAT3 synergistically controlled MDA-MB-231 cell migration by recruiting DNMT1 to hypermethylate the promoter of BRMS1 and thus affect the expression of BRMS1. Interestingly, physical interaction between MRTF-A and STAT3 synergistically promotes the transactivity of DNMT1 by binding to the GAS element within the DNMT1 promoter. Our data thus provide important and novel insights into the roles of MRTF-A and STAT3 in regulating MDA-MB-231 cell migration.


Subject(s)
Breast Neoplasms/pathology , Repressor Proteins/genetics , STAT3 Transcription Factor/metabolism , Trans-Activators/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Humans , Osteopontin/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , STAT3 Transcription Factor/genetics , Trans-Activators/genetics , Urokinase-Type Plasminogen Activator/metabolism
5.
Cell Signal ; 26(12): 2738-48, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25152367

ABSTRACT

Myocardin is well known to play a key role in the development of cardiomyocyte hypertrophy. But the exact molecular mechanism regulating myocardin stability and transactivity to affect cardiomyocyte hypertrophy has not been studied clearly. We now report that NF-κB (p65) can inhibit myocardin-induced cardiomyocyte hypertrophy. Then we explore the molecular mechanism of this response. First, we show that p65 can functionally repress myocardin transcriptional activity and also reduce the protein expression of myocardin. Second, the function of myocardin can be regulated by epigenetic modifications. Myocardin sumoylation is known to transactivate cardiac genes, but whether p65 can inhibit SUMO modification of myocardin is still not clear. Our data show that p65 weakens myocardin transcriptional activity through attenuating SUMO modification of myocardin by SUMO1/PIAS1, thereby impairing myocardin-mediated cardiomyocyte hypertrophy. Furthermore, the expression of myocardin can be regulated by several microRNAs, which play important roles in the development and function of the heart and muscle. We next investigated potential role of miR-1 in cardiac hypotrophy. Our results show that p65 can upregulate the level of miR-1 and miR-1 can decrease protein expression of myocardin in cardiac myocytes. Notably, miR-1 expression is also controlled by myocardin, leading to a feedback loop. These data thus provide important and novel insights into the function that p65 inhibits myocardin-mediated cardiomyocyte hypertrophy by downregulating the expression and SUMO modification of myocardin and enhancing the expression of miR-1.


Subject(s)
Hypertrophy/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , Down-Regulation/physiology , MicroRNAs/metabolism , Protein Inhibitors of Activated STAT/metabolism , Rats , Rats, Sprague-Dawley , SUMO-1 Protein/metabolism , Transcription, Genetic/physiology , Transcriptional Activation/physiology , Up-Regulation/physiology
6.
Cell Signal ; 26(11): 2370-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25038455

ABSTRACT

Breast cancer is the leading cause of cancer death in women worldwide which is closely related to metastasis. But the exact molecular mechanism on metastasis is still not fully understood; we now report that both MRTF-A and STAT3 play important role in breast cancer migration of MDA-MB-231 cells. Moreover, MRTF-A and STAT3 synergistically increased MDA-MB-231 cell migration by promoting the expression of migration markers Myl-9 and Cyr-61. Importantly, we identified a detailed molecular mechanism of MDA-MB-231 cell migration controlled via physical interaction between MRTF-A and STAT3, which synergistically promote the transactivity of the migration marker Myl-9 and Cyr-61 by CArG box binding. Interestingly, the two signaling pathways RhoA-MRTF-A and JAK-STAT3 across talk to regulate MDA-MB-231 cell migration. Our data thus provide important and novel insights into the roles of MRTF-A and STAT3 in regulating MDA-MB-231 cell migration.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , STAT3 Transcription Factor/biosynthesis , Trans-Activators/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cysteine-Rich Protein 61/biosynthesis , Cysteine-Rich Protein 61/genetics , Female , Humans , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Neoplasm Proteins/genetics , Response Elements , STAT3 Transcription Factor/genetics , Trans-Activators/genetics , Transcriptional Activation/genetics
7.
IUBMB Life ; 66(5): 352-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24753159

ABSTRACT

Human chorionic gonadotropin (hCG) is a glycoprotein produced by placental trophoblasts. Previous studies indicated that hCG could be responsible for the pregnancy-induced protection against breast cancer in women. It is reported that hCG decreases proliferation and invasion of breast cancer MCF-7 cells. Our research also demonstrates that hCG can reduce the proliferation of MCF-7 cells by downregulating the expression of proliferation markers, proliferating cell nuclear antigen (PCNA), and proliferation-related Ki-67 antigen (Ki-67). Interestingly, we find here that hCG elevates the state of cellular differentiation, as characterized by the upregulation of differentiation markers, ß-casein, cytokeratin-18 (CK-18), and E-cadherin. Inhibition of hCG secretion or luteinizing hormone/hCG receptors (LH/hCGRs) synthesis can weaken the effect of hCG on the induction of cell differentiation. Furthermore, hCG can suppress the expression of estrogen receptor alpha. hCG activated receptor-mediated cyclic adenosine monophosphate/protein kinase A signaling pathway. These findings indicated that a protective effect of hCG against breast cancer may be associated with its growth inhibitory and differentiation induction function in breast cancer cells.


Subject(s)
Cell Proliferation , Chorionic Gonadotropin/physiology , Antigens, CD , Breast Neoplasms , Cadherins/metabolism , Caseins/metabolism , Cell Differentiation , Cyclic AMP/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Heparan Sulfate Proteoglycans/metabolism , Humans , Keratin-18/metabolism , MCF-7 Cells , Receptors, LH/genetics , Receptors, LH/metabolism , Second Messenger Systems
8.
Cell Signal ; 26(6): 1335-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24607789

ABSTRACT

Breast cancer is the leading cause of cancer death in women worldwide. It is well known that oncogene activation and anti-oncogene inactivation affect the development and progression of breast cancer, but the role of oncogene activation and anti-oncogene inactivation in breast cancer is still not fully understood. We now report that maspin acts as a tumor suppressor gene to induce MCF-7 cell apoptosis. In addition, maspin promoter hypermethylation and histone hypoacetylation lead to silencing of maspin gene expression in MCF-7 cells. Moreover, DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-aza-dc) and/or the histone deacetylase (HDAC) inhibitor Trichostatin A (TSA) strongly up-regulated the expression of maspin in MCF-7 cells. Notably, myocardin can promote the re-expression of maspin in MCF-7 cells. Luciferase assay shows that myocardin activates the transcription of maspin promoter by CArG box. More importantly, 5-aza-dc/TSA and myocardin synergetically enhance re-expression of maspin and augment maspin-mediated apoptosis in MCF-7 cells. Thus, these data reveal the new insight that myocardin meditates apoptosis in breast cancer through affecting maspin re-expression and epigenetic modification to regulate the development of breast cancer, thereby raising the possibility of its use in breast cancer therapy.


Subject(s)
Epigenesis, Genetic , Nuclear Proteins/physiology , Serpins/genetics , Trans-Activators/physiology , Transcription, Genetic , Acetylation , Apoptosis , Base Sequence , Breast Neoplasms , Cell Proliferation , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Histones/metabolism , Humans , MCF-7 Cells , Promoter Regions, Genetic , Protein Processing, Post-Translational , Serpins/metabolism
9.
Clin Exp Pharmacol Physiol ; 40(1): 13-21, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23110505

ABSTRACT

The aim of the present study was to analyse the antiproliferative effects and mechanisms of action of protein kinase inhibitors (PKIs) in human glioblastoma multiforme (GBM) cells with different epidermal growth factor receptor (EGFR) and phosphatase and tensin homologue (PTEN) status. The GBM cell models were established by transfection of plasmids carrying wild-type EGFR, mutated EGFRvIII or PTEN and clonal selection in U87MG cells. Phosphatidylinositol 3-kinase (PI3-K)/AKT pathway-focused gene profiles were examined by real-time polymerase chain reaction-based assays, protein expression was evaluated by western blotting and the antiproliferative effects of PKI treatment were determined by the 3-(4,5-dimethyl-2 thiazoyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay in GBM cells. The cell model with intact PTEN and low EGFR levels was the most sensitive to treatment with the EGFR inhibitor erlotinib, whereas the model with EGFRvIII was the most resistant to treatment with the mitogen-activated protein kinase kinase inhibitor U0126. The dual PI3-K and mammalian target of rapamycin (mTOR) inhibitor PI103 had the most potent antiproliferative effects against all GBM cells tested. Following simultaneous stimulation of AKT and extracellular signal-regulated kinase, rapamycin concentrations > 0.5 nmol/L failed to exhibit a further growth inhibitory effect. Concurrent inhibition of mTOR and ribosomal protein s6 activity may underlie the inhibition of GBM proliferation by PKI. In conclusion, overexpression of EGFR or EGFRvIII, accompanied by a loss of PTEN, contributed to the activation of multiple intracellular signalling pathways in GBM cells. Rigorous examination of biomarkers in tumour tissues before and after treatment may be necessary to determine the efficacy of PKI therapy in patients with GBM.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/drug therapy , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/biosynthesis , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Furans/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Nitriles/pharmacology , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
10.
Mol Cell Biochem ; 366(1-2): 345-54, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22527939

ABSTRACT

Acute myocardial infarction (AMI) is a leading cause of death worldwide. Most cases of AMI result from coronary atherosclerosis (AS). The pathogenic mechanisms underlying AS lesions and AMI are incompletely understood. Calcium-sensing receptors (CaSR) belong to a family of G-protein-coupled receptors. We previously discovered that CaSR was expressed in the heart tissue of adult rats. CaSR may contribute to AMI in AS. We initially established a rat model of AS by injection of vitamin D(3) and feeding with a high-fat diet. Isoproterenol (ISO) was used to induce AMI. The MB isoenzyme of creatine kinase (CK-MB), lactate dehydrogenase (LDH), cardiac troponin T (cTnT), tetrazolium chloride staining, and cardiac function parameters were selected as indicators of myocardial damage or necrosis. Cardiac apoptosis was analyzed by transferase dUTP nick-end labeling (TUNEL) assay. Expression of CaSR, Bcl-2, Bax, caspase-3, p-ERK1/2, p-JNK, and p-p38 were determined by Western blot analysis. Compared with the control group, levels of cTnT, CK-MB, and LDH; number of TUNEL-positive cells; and expression of CaSR, Bax, caspase-3, p-ERK1/2, p-JNK and p-p38, were significantly increased, whereas cardiac function and expression of Bcl-2 were decreased markedly in isoproterenol (ISO)-treated group (C/ISO) and AS groups. These changes were significant in the AS/ISO group than in the C/ISO group or AS group. The upregulation of CaSR during AS formation renders hypersensitivity to AMI. Activation of the pro-apoptotic mitochondria pathway and JNK-p38 MAPK pathway triggered by increased expression of CaSR may be one of molecular mechanisms underlying AMI in AS.


Subject(s)
Atherosclerosis/metabolism , Myocardial Infarction/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Aorta, Abdominal/pathology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Atherosclerosis/blood , Atherosclerosis/etiology , Cholesterol/blood , Creatine Kinase, MB Form/blood , Diet, High-Fat/adverse effects , Disease Susceptibility , Isoproterenol , L-Lactate Dehydrogenase/blood , Male , Mitogen-Activated Protein Kinases/metabolism , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Wistar , Receptors, Calcium-Sensing/genetics , Triglycerides/blood , Troponin T/metabolism , Up-Regulation , Ventricular Function
11.
Mol Cell Biochem ; 362(1-2): 115-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22083546

ABSTRACT

Matrix metalloproteinase-2 (MMP-2) is constitutively expressed in vascular smooth muscle cells (VSMCs) and up-regulated in atherosclerotic lesion by various stimuli, such as oxidized low-density lipoprotein (oxLDL). Calcium-sensing receptor (CaSR) is also expressed in VSMCs, but it remains unclear whether CaSR is associated with overproduction of MMP-2 in VSMCs. In this study, the expression of MMP-2 was detected by real-time PCR and Western blot analysis, and the gelatinolytic activity of MMP-2 was measured using gelatin zymography. Our results showed that oxLDL enhanced MMP-2 expression and activity in rat aortic VSMCs in a time- and dose-dependent manner. In addition, CaSR expression was up-regulated by oxLDL. Manipulating CaSR function in these cells by NPS2390 (an antagonist of CaSR) or GdCl(3) (an agonist of CaSR) affected the oxLDL-induced MMP-2 production. In VSMCs, oxLDL stimulated the rapid activation of phosphatidylinositol 3-kinase (PI3K)/Akt signal pathway, as determined by Western blot analysis. Phosphorylation of Akt and MMP-2 production stimulated by oxLDL were attenuated by LY294002 (a specific inhibitor of PI3K). Activation of Akt was suppressed by NPS2390 but enhanced by GdCl(3). In contrast, oxLDL had no stimulatory effect on the phosphorylation of JNK, and pretreatment with SP600125 (an inhibitor of JNK) produced no significant effect on oxLDL-induced MMP-2 production. These results suggest that CaSR mediates oxLDL-induced MMP-2 production in VSMCs via PI3K/Akt signal pathway.


Subject(s)
Lipoproteins, LDL/metabolism , Matrix Metalloproteinase 2/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Calcium-Sensing/metabolism , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Anthracenes/pharmacology , Aorta/metabolism , Atherosclerosis/metabolism , Cells, Cultured , Chromones/pharmacology , Gadolinium/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Male , Matrix Metalloproteinase 2/metabolism , Morpholines/pharmacology , Muscle, Smooth, Vascular/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/biosynthesis
12.
Clin Exp Pharmacol Physiol ; 38(9): 605-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21692826

ABSTRACT

1. Calcium-sensing receptors (CaSR) exist in a variety of tissues. In 2010, we first identified its functional expression in Buffalo rat liver (BRL) cells and demonstrated that the activation of CaSR was involved in an increased intracellular calcium through the Gq subunit-phospholipase C-inositol triphosphate pathway. However, its role and related mechanism in hepatic ischaemia/reperfusion (I/R) injury is still unclear. 2. Therefore, in the present study, BRL cells were incubated in ischaemia-mimetic solution for 4 h, then reincubated in the normal culture medium for 10 h to establish a simulated I/R model. We assayed the apoptotic ratio of BRL cells by flow cytometry and Hoechst 33342 staining; analyzed the expression of CaSR, cytochrome c (Cyt-c), caspase-3, Bcl-2, Bax, extracellular signal-regulated protein kinase (ERK), and p38 by Western blotting; and measured the concentration of intracellular calcium by laser-scanning confocal microscopy. 3. The results showed that simulated I/R increased the expression of CaSR and induced apoptosis in BRL cells. GdCl(3), a specific activator of CaSR, further increased CaSR expression, intracellular calcium, and apoptosis in BRL cells during I/R. The activation of CaSR downregulated Bcl-2 expression, upregulated Cyt-c, caspase-3, and Bax expressions, and promoted p38 and ERK-1/2 phosphorylation. 4. In conclusion, increased CaSR expression plays a vital role in apoptosis induced by I/R injury, in which its mechanism is related with calcium overload and the activation of the mitochondrial and mitogen-activated protein kinase apoptotic pathways. The regulation of CaSR activity might serve as a novel pharmacological target to prevent and treat liver disease.


Subject(s)
Apoptosis/physiology , Hepatocytes/cytology , Hepatocytes/metabolism , Receptors, Calcium-Sensing/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Animals , Apoptosis/genetics , Calcium/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cells, Cultured , Cytochromes c/genetics , Cytochromes c/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Inositol Polyphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Rats , Rats, Inbred BUF , Receptors, Calcium-Sensing/genetics , Reperfusion Injury/genetics , Signal Transduction/physiology , Type C Phospholipases/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Biomed Sci ; 18: 16, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21314926

ABSTRACT

BACKGROUND: The extracellular calcium-sensing receptor (CaSR) belongs to family C of the G protein coupled receptors. Whether the CaSR is expressed in the pulmonary artery (PA) is unknown. METHODS: The expression and distribution of CaSR were detected by RT-PCR, Western blotting and immunofluorescence. PA tension was detected by the pulmonary arterial ring technique, and the intracellular calcium concentration ([Ca2+]i) was detected by a laser-scanning confocal microscope. RESULTS: The expressions of CaSR mRNA and protein were found in both rat pulmonary artery smooth muscle cells (PASMCs) and PAs. Increased levels of [Ca2+]o (extracellular calcium concentration) or Gd3+ (an agonist of CaSR) induced an increase of [Ca2+]i and PAs constriction in a concentration-dependent manner. In addition, the above-mentioned effects of Ca2+ and Gd3+ were inhibited by U73122 (specific inhibitor of PLC), 2-APB (specific antagonist of IP3 receptor), and thapsigargin (blocker of sarcoplasmic reticulum calcium ATPase). CONCLUSIONS: CaSR is expressed in rat PASMCs, and is involved in regulation of PA tension by increasing [Ca2+]i through G-PLC-IP3 pathway.


Subject(s)
Pulmonary Artery/metabolism , Receptors, Calcium-Sensing/genetics , Animals , Base Sequence , Blotting, Western , Boron Compounds/pharmacology , Calcium/pharmacology , Calcium Channel Blockers/pharmacology , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Fluorescent Antibody Technique , In Vitro Techniques , Male , Molecular Sequence Data , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Pyrrolidinones/pharmacology , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Calcium-Sensing/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thapsigargin/pharmacology , Type C Phospholipases/antagonists & inhibitors
14.
Basic Clin Pharmacol Toxicol ; 108(3): 185-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21073657

ABSTRACT

Activation of the calcium-sensing receptor (CaSR) leads to an increase of intracellular calcium concentration and alteration of cellular activities. High level of intracellular calcium is involved in hypoxia-induced proliferation of pulmonary arterial smooth muscle cells (PASMCs). However, whether the CaSR is expressed in PAMSCs and is related to the hypoxia-induced proliferation of PASMCs is unclear. In this study, the expression and distribution of CaSRs were detected by RT-PCR, western blotting and immunofluorescence; the intracellular concentration of free calcium ([Ca(2+) ](i) ) was determined by confocal laser scanning microscopy; cell proliferation was tested using an MTT and BrdU incorporation assay; cell cycle analysis was carried out using a flow cytometric assay; and the expression of proliferating cell nuclear antigen (PCNA), extracellular signal-regulated protein kinase 1,2 (ERK1,2) and AKT were analysed by western blotting. We observed that both CaSR mRNA and protein were expressed in rat PASMCs. Lowering of oxygen from 21% to 2.5% led to increased [Ca(2+) ](i) and CaSR expression. This condition of hypoxia also stimulated PASMCs proliferation accompanying with increased phosphorylation of ERK1,2 and AKT. GdCl(3) (an agonist of CaSR) or NPS2390 (an antagonist of CaSR) amplified or weakened the effect of hypoxia, respectively. PD98059 (a MEK1 inhibitor) or LY294002 (a PI3K inhibitors) decreased the up-regulation of PCNA expression and the increase of the cell proliferation index induced by hypoxia and GdCl(3) in PASMCs. Our results suggest that CaSR is expressed in rat PASMCs, and that CaSR activation through MEK1/ERK1,2 and PI3 kinase pathways is involved in hypoxia-induced proliferation of PASMCs.


Subject(s)
Cell Hypoxia , Cell Proliferation , MAP Kinase Signaling System , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Pulmonary Artery/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Calcium Signaling/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/antagonists & inhibitors , Receptors, Calcium-Sensing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...