Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(6)2023 06 06.
Article in English | MEDLINE | ID: mdl-37371532

ABSTRACT

An elevated level of circulating homocysteine (Hcy) has been regarded as an independent risk factor for cardiovascular disease; however, the clinical benefit of Hcy lowering-therapy is not satisfying. To explore potential unrevealed mechanisms, we investigated the roles of Ca2+ influx through TRPC channels and regulation by Hcy-copper complexes. Using primary cultured human aortic endothelial cells and HEK-293 T-REx cells with inducible TRPC gene expression, we found that Hcy increased the Ca2+ influx in vascular endothelial cells through the activation of TRPC4 and TRPC5. The activity of TRPC4 and TRPC5 was regulated by extracellular divalent copper (Cu2+) and Hcy. Hcy prevented channel activation by divalent copper, but monovalent copper (Cu+) had no effect on the TRPC channels. The glutamic acids (E542/E543) and the cysteine residue (C554) in the extracellular pore region of the TRPC4 channel mediated the effect of Hcy-copper complexes. The interaction of Hcy-copper significantly regulated endothelial proliferation, migration, and angiogenesis. Our results suggest that Hcy-copper complexes function as a new pair of endogenous regulators for TRPC channel activity. This finding gives a new understanding of the pathogenesis of hyperhomocysteinemia and may explain the unsatisfying clinical outcome of Hcy-lowering therapy and the potential benefit of copper-chelating therapy.


Subject(s)
Copper , Endothelial Cells , Humans , Copper/pharmacology , Copper/metabolism , Endothelial Cells/metabolism , HEK293 Cells , Carrier Proteins , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Calcium/metabolism
2.
Life Sci ; 306: 120834, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-35902031

ABSTRACT

Mitochondria play a pivotal role in cellular function, not only acting as the powerhouse of the cell, but also regulating ATP synthesis, reactive oxygen species (ROS) production, intracellular Ca2+ cycling, and apoptosis. During the past decade, extensive progress has been made in the technology to assess mitochondrial functions and accumulating evidences have shown that mitochondrial dysfunction is a key pathophysiological mechanism for many diseases including cardiovascular disorders, such as ischemic heart disease, cardiomyopathy, hypertension, atherosclerosis, and hemorrhagic shock. The advances in methodology have been accelerating our understanding of mitochondrial molecular structure and function, biogenesis and ROS and energy production, which facilitates new drug target identification and therapeutic strategy development for mitochondrial dysfunction-related disorders. This review will focus on the assessment of methodologies currently used for mitochondrial research and discuss their advantages, limitations and the implications of mitochondrial dysfunction in cardiovascular disorders.


Subject(s)
Cardiovascular Diseases , Mitochondrial Diseases , Apoptosis , Cardiovascular Diseases/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
3.
PLoS One ; 16(2): e0247234, 2021.
Article in English | MEDLINE | ID: mdl-33606763

ABSTRACT

Elevated reactive oxygen species (ROS) in type 2 diabetes cause cellular damage in many organs. Recently, the new class of glucose-lowering agents, SGLT-2 inhibitors, have been shown to reduce the risk of developing diabetic complications; however, the mechanisms of such beneficial effect are largely unknown. Here we aimed to investigate the effects of dapagliflozin on cell proliferation and cell death under oxidative stress conditions and explore its underlying mechanisms. Human proximal tubular cells (HK-2) were used. Cell growth and death were monitored by cell counting, water-soluble tetrazolium-1 (WST-1) and lactate dehydrogenase (LDH) assays, and flow cytometry. The cytosolic and mitochondrial (ROS) production was measured using fluorescent probes (H2DCFDA and MitoSOX) under normal and oxidative stress conditions mimicked by addition of H2O2. Intracellular Ca2+ dynamics was monitored by FlexStation 3 using cell-permeable Ca2+ dye Fura-PE3/AM. Dapagliflozin (0.1-10 µM) had no effect on HK-2 cell proliferation under normal conditions, but an inhibitory effect was seen at an extreme high concentration (100 µM). However, dapagliflozin at 0.1 to 5 µM showed remarkable protective effects against H2O2-induced cell injury via increasing the viable cell number at phase G0/G1. The elevated cytosolic and mitochondrial ROS under oxidative stress was significantly decreased by dapagliflozin. Dapagliflozin increased the basal intracellular [Ca2+]i in proximal tubular cells, but did not affect calcium release from endoplasmic reticulum and store-operated Ca2+ entry. The H2O2-sensitive TRPM2 channel seemed to be involved in the Ca2+ dynamics regulated by dapagliflozin. However, dapagliflozin had no direct effects on ORAI1, ORAI3, TRPC4 and TRPC5 channels. Our results suggest that dapagliflozin shows anti-oxidative properties by reducing cytosolic and mitochondrial ROS production and altering Ca2+ dynamics, and thus exerts its protective effects against cell damage under oxidative stress environment.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Kidney Tubules, Proximal/pathology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cytosol/metabolism , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/genetics , Phenanthridines/metabolism , Protein Isoforms/metabolism , Reactive Oxygen Species/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2/metabolism
4.
Expert Opin Drug Discov ; 16(4): 447-461, 2021 04.
Article in English | MEDLINE | ID: mdl-33003971

ABSTRACT

INTRODUCTION: Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease (ESRD), and 40% of patients with diabetes develop DKD. Although some pathophysiological mechanisms and drug targets of DKD have been described, the effectiveness or clinical usefulness of such treatment has not been well validated. Therefore, searching for new targets and potential therapeutic candidates has become an emerging research area. AREAS COVERED: The pathophysiological mechanisms, new drug targets and potential therapeutic compounds for DKD are addressed in this review. EXPERT OPINION: Although preclinical and clinical evidence has shown some positive results for controlling DKD progression, treatment regimens have not been well developed to reduce the mortality in patients with DKD globally. Therefore, the discovery of new therapeutic targets and effective target-based drugs to achieve better and safe treatment are urgently required. Preclinical screening and clinical trials for such drugs are needed.


Subject(s)
Diabetic Nephropathies/drug therapy , Drug Discovery/methods , Kidney Failure, Chronic/prevention & control , Animals , Diabetic Nephropathies/complications , Diabetic Nephropathies/physiopathology , Disease Progression , Drug Development , Humans , Kidney Failure, Chronic/etiology
5.
Stem Cell Res Ther ; 11(1): 395, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32928296

ABSTRACT

BACKGROUND: Bone mesenchymal stem cells (MSCs) can promote liver regeneration and inhibit inflammation and hepatic fibrosis. MSCs also can serve as a vehicle for gene therapy. Smad7 is an essential negative regulatory gene in the TGF-ß1/Smad signalling pathway. Activation of TGF-ß1/Smad signalling accelerates liver inflammation and fibrosis; we therefore hypothesized that MSCs overexpressing the Smad7 gene might be a new cell therapy approach for treating liver fibrosis via the inhibition of TGF-ß1/Smad signalling. METHODS: MSCs were isolated from 6-week-old Wistar rats and transduced with the Smad7 gene using a lentivirus vector. Liver cirrhosis was induced by subcutaneous injection of carbon tetrachloride (CCl4) for 8 weeks. The rats with established liver cirrhosis were treated with Smad7-MSCs by direct injection of cells into the main lobes of the liver. The expression of Smad7, Smad2/3 and fibrosis biomarkers or extracellular matrix proteins and histopathological change were assessed by quantitative PCR, ELISA and Western blotting and staining. RESULTS: The mRNA and protein level of Smad7 in the recipient liver and serum were increased after treating with Smad-MSCs for 7 and 21 days (P < 0.001). The serum levels of collagen I and III and collagenase I and III were significantly (P < 0.001) reduced after the treatment with Smad7-MSCs. The mRNA levels of TGF-ß1, TGFBR1, α-SMA, TIMP-1, laminin and hyaluronic acid were decreased (P < 0.001), while MMP-1 increased (P < 0.001). The liver fibrosis score and liver function were significantly alleviated after the cell therapy. CONCLUSIONS: The findings suggest that the MSC therapy with Smad7-MSCs is effective in the treatment of liver fibrosis in the CCl4-induced liver cirrhosis model. Inhibition of TGF-ß1 signalling pathway by enhancement of Smad-7 expression could be a feasible cell therapy approach to mitigate liver cirrhosis.


Subject(s)
Liver Cirrhosis, Experimental , Mesenchymal Stem Cells , Animals , Genetic Therapy , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Liver Cirrhosis, Experimental/pathology , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Smad7 Protein/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
6.
J Am Soc Nephrol ; 30(10): 1910-1924, 2019 10.
Article in English | MEDLINE | ID: mdl-31416818

ABSTRACT

BACKGROUND: Mutations in the transient receptor potential channel 6 (TRPC6) gene are associated with an inherited form of FSGS. Despite widespread expression, patients with TRPC6 mutations do not present with any other pathologic phenotype, suggesting that this protein has a unique yet unidentified role within the target cell for FSGS, the kidney podocyte. METHODS: We generated a stable TRPC6 knockout podocyte cell line from TRPC6 knockout mice. These cells were engineered to express wild-type TRPC6, a dominant negative TRPC6 mutation, or either of two disease-causing mutations of TRPC6, G109S or K874*. We extensively characterized these cells using motility, detachment, and calpain activity assays; immunofluorescence; confocal or total internal reflection fluorescence microscopy; and western blotting. RESULTS: Compared with wild-type cells, TRPC6-/- podocytes are less motile and more adhesive, with an altered actin cytoskeleton. We found that TRPC6 binds to ERK1/2 and the actin regulatory proteins, caldesmon (a calmodulin- and actin-binding protein) and calpain 1 and 2 (calcium-dependent cysteine proteases that control the podocyte cytoskeleton, cell adhesion, and motility via cleavage of paxillin, focal adhesion kinase, and talin). Knockdown or expression of the truncated K874* mutation (but not expression of the gain-of-function G019S mutation or dominant negative mutant of TRPC6) results in the mislocalization of calpain 1 and 2 and significant downregulation of calpain activity; this leads to altered podocyte cytoskeleton, motility, and adhesion-characteristics of TRPC6-/- podocytes. CONCLUSIONS: Our data demonstrate that independent of TRPC6 channel activity, the physical interaction between TRPC6 and calpain in the podocyte is important for cell motility and detachment and demonstrates a scaffolding role of the TRPC6 protein in disease.


Subject(s)
Calpain/physiology , Cell Adhesion , Cell Movement , Cytoskeleton/physiology , Podocytes/physiology , Podocytes/ultrastructure , TRPC6 Cation Channel/physiology , Animals , Mice , Mice, Knockout
7.
Br J Pharmacol ; 176(19): 3845-3856, 2019 10.
Article in English | MEDLINE | ID: mdl-31271653

ABSTRACT

BACKGROUND AND PURPOSE: Mibefradil, a T-type Ca2+ channel blocker, has been investigated for treating solid tumours. However, its underlying mechanisms are still unclear. Here, we have investigated the pharmacological actions of mibefradil on Orai store-operated Ca2+ channels. EXPERIMENTAL APPROACH: Human Orai1-3 cDNAs in tetracycline-regulated pcDNA4/TO vectors were transfected into HEK293 T-REx cells with stromal interaction molecule 1 (STIM1) stable expression. The Orai currents were recorded by whole-cell and excised-membrane patch clamp. Ca2+ influx or release was measured by Fura-PE3/AM. Cell growth and death were monitored by WST-1, LDH assays and flow cytometry. KEY RESULTS: Mibefradil inhibited Orai1, Orai2, and Orai3 currents dose-dependently. The IC50 for Orai1, Orai2, and Orai3 channels was 52.6, 14.1, and 3.8 µM respectively. Outside-out patch demonstrated that perfusion of 10-µM mibefradil to the extracellular surface completely blocked Orai3 currents and single channel activity evoked by 2-APB. Intracellular application of mibefradil did not alter Orai3 channel activity. Mibefradil at higher concentrations (>50 µM) inhibited Ca2+ release but had no effect on cytosolic STIM1 translocation evoked by thapsigargin. Inhibition on Orai channels by mibefradil was structure-related, as other T-type Ca2+ channel blockers with different structures, such as ethosuximide and ML218, had no or minimal effects on Orai channels. Moreover, mibefradil inhibited cell proliferation, induced apoptosis, and arrested cell cycle progression. CONCLUSIONS AND IMPLICATIONS: Mibefradil is a potent cell surface blocker of Orai channels, demonstrating a new pharmacological action of this compound in regulating cell growth and death, which could be relevant to its anti-cancer activity.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Mibefradil/pharmacology , ORAI1 Protein/antagonists & inhibitors , ORAI2 Protein/antagonists & inhibitors , Calcium/analysis , Calcium/metabolism , Calcium Channel Blockers/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Cells, Cultured , HEK293 Cells , Humans , Mibefradil/chemistry , ORAI1 Protein/metabolism , ORAI2 Protein/metabolism , Optical Imaging
8.
Nat Commun ; 8(1): 1920, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203863

ABSTRACT

Impaired albumin reabsorption by proximal tubular epithelial cells (PTECs) has been highlighted in diabetic nephropathy (DN), but little is known about the underlying molecular mechanisms. Here we find that ORAI1-3, are preferentially expressed in PTECs and downregulated in patients with DN. Hyperglycemia or blockade of insulin signaling reduces the expression of ORAI1-3. Inhibition of ORAI channels by BTP2 and diethylstilbestrol or silencing of ORAI expression impairs albumin uptake. Transgenic mice expressing a dominant-negative Orai1 mutant (E108Q) increases albuminuria, and in vivo injection of BTP2 exacerbates albuminuria in streptozotocin-induced and Akita diabetic mice. The albumin endocytosis is Ca2+-dependent and accompanied by ORAI1 internalization. Amnionless (AMN) associates with ORAIs and forms STIM/ORAI/AMN complexes after Ca2+ store depletion. STIM1/ORAI1 colocalizes with clathrin, but not with caveolin, at the apical membrane of PTECs, which determines clathrin-mediated endocytosis. These findings provide insights into the mechanisms of protein reabsorption and potential targets for treating diabetic proteinuria.


Subject(s)
Albumins/metabolism , Albuminuria/genetics , Calcium Channels/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , ORAI1 Protein/genetics , ORAI2 Protein/genetics , Albumins/drug effects , Albuminuria/metabolism , Anilides/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers , Calcium Channels/metabolism , Case-Control Studies , Caveolins/metabolism , Cell Line , Clathrin/metabolism , Diethylstilbestrol/pharmacology , Down-Regulation , Endocytosis , Epithelial Cells/drug effects , Estrogens, Non-Steroidal/pharmacology , Female , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Kidney/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Middle Aged , Mutation , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/metabolism , ORAI2 Protein/antagonists & inhibitors , ORAI2 Protein/metabolism , Renal Reabsorption/drug effects , Renal Reabsorption/genetics , Stromal Interaction Molecule 1/metabolism , Thiadiazoles/pharmacology
9.
J Mol Med (Berl) ; 93(5): 511-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25471481

ABSTRACT

UNLABELLED: ORAI and stromal interaction molecule (STIM) are store-operated channel molecules that play essential roles in human physiology through a coupling mechanism of internal Ca(2+) store to Ca(2+) influx. However, the roles of ORAI and STIM in vascular endothelial cells under diabetic conditions remain unknown. Here, we investigated expression and signalling pathways of ORAI and STIM regulated by high glucose or hyperglycaemia using in vitro cell models, in vivo diabetic mice and tissues from patients. We found that ORAI1-3 and STIM1-2 were ubiquitously expressed in human vasculatures. Their expression was upregulated by chronic treatment with high glucose (HG, 25 mM D-glucose), which was accompanied by enhanced store-operated Ca(2+) influx in vascular endothelial cells. The increased expression was also observed in the aortae from genetically modified Akita diabetic mice (C57BL/6-Ins2(Akita)/J) and streptozocin-induced diabetic mice, and aortae from diabetic patients. HG-induced upregulation of ORAI and STIM genes was prevented by the calcineurin inhibitor cyclosporin A and NFATc3 siRNA. Additionally, in vivo treatment with the nuclear factor of activated T cells (NFAT) inhibitor A-285222 prevented the gene upregulation in Akita mice. However, HG had no direct effects on ORAI1-3 currents and the channel activation process through cytosolic STIM1 movement in the cells co-expressing STIM1-EYFP/ORAIs. We concluded that upregulation of STIM/ORAI through Ca(2+)-calcineurin-NFAT pathway is a novel mechanism causing abnormal Ca(2+) homeostasis and endothelial dysfunction under hyperglycaemia. KEY MESSAGE: ORAI1-3 and STIM1-2 are ubiquitously expressed in vasculatures and upregulated by high glucose. Increased expression is confirmed in Akita (Ins2(Akita)/J) and STZ diabetic mice and patients. Upregulation mechanism is mediated by Ca(2+)/calcineurin/NFATc3 signalling. High glucose has no direct effects on ORAI1-3 channel activity and channel activation process.


Subject(s)
Calcineurin/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Glucose/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction , Animals , Calcium Channels/genetics , Cell Line , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Middle Aged , RNA, Messenger , Up-Regulation
10.
Br J Pharmacol ; 171(5): 1250-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24670147

ABSTRACT

BACKGROUND: Depletion of the Ca(2+) store by ryanodine receptor (RyR) agonists induces store-operated Ca(2+) entry (SOCE). 4-Chloro-3-ethylphenol (4-CEP) and 4-chloro-m-cresol (4-CmC) are RyR agonists commonly used as research tools and diagnostic reagents for malignant hyperthermia. Here, we investigated the effects of 4-CEP and its analogues on SOCE. EXPERIMENTAL APPROACH: SOCE and ORAI1-3 currents were recorded by Ca(2+) imaging and whole-cell patch recordings in rat L6 myoblasts and in HEK293 cells overexpressing STIM1/ORAI1-3. KEY RESULTS: 4-CEP induced a significant release of Ca(2+) in rat L6 myoblasts, but inhibited SOCE. The inhibitory effect was concentration-dependent and more potent than its analogues 4-CmC and 4-chlorophenol (4-ClP). In the HEK293 T-REx cells overexpressing STIM1/ORAI1-3, 4-CEP inhibited the ORAI1, ORAI2 and ORAI3 currents evoked by thapsigargin. The 2-APB-induced ORAI3 current was also blocked by 4-CEP. This inhibitory effect was reversible and independent of the Ca(2+) release. The two analogues, 4-CmC and 4-ClP, also inhibited the ORAI1-3 channels. Excised patch and intracellular application of 4-CEP demonstrated that the action site was located extracellularly. Moreover, 4-CEP evoked STIM1 translocation and subplasmalemmal clustering through its Ca(2+) store-depleting effect via the activation of RyR, but no effect on STIM1 redistribution was observed in cells co-expressing STIM1/ORAI1-3. CONCLUSION AND IMPLICATIONS: 4-CEP not only acts as a RyR agonist to deplete the Ca(2+) store and trigger STIM1 subplasmalemmal translocation and clustering, but also directly inhibits ORAI1-3 channels. These findings demonstrate a novel pharmacological property for the chlorophenol derivatives that act as RyR agonists.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channels/metabolism , Chlorophenols/pharmacology , Cresols/pharmacology , Membrane Proteins/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Channels/genetics , Cell Line , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , ORAI2 Protein , Rats , Ryanodine Receptor Calcium Release Channel/metabolism , Stromal Interaction Molecule 1
11.
Mol Biol Rep ; 41(4): 2345-51, 2014.
Article in English | MEDLINE | ID: mdl-24415303

ABSTRACT

Bovine mastitis is a very complex and common disease of dairy cattle and a major source of economic losses to the dairy industry worldwide. In this study, the bovine TLR4 was taken as a candidate gene for mastitis resistance. This study aimed to analyze the associations of single nucleotide polymorphisms (SNP) or haplotype and somatic cell score (SCS) in 404 Chinese commercial dairy cattle including Chinese Holstein, Sanhe cattle and Chinese Simmental breeds. The polymerase chain reaction and sequencing methods were used for detecting genotype and allele frequency distribution of the two SNPs (rs8193062, rs8193064), statistical results showed that T allele at rs8193062 and C allele at rs8193064 were the predominate alleles. Moreover, six SNPs, including two SNPs (rs8193062, rs8193064) and four SNPs (rs8193060, rs8193069, rs29017188, rs8193046) which were chosen according the polymorphism level for the same cattle populations in previous studies, were used for haplotype analysis, the results revealed that twenty-one haplotypes were found in the mentioned animals, of which, Hap1 (30.5 %) and Hap2 (30.4 %) were the most common haplotypes. Hap2, Hap4 and Hap12 might negatively effect on milk SCS, whereas Hap13 might positively effect on milk SCS. The results in this study might assist in marker assisted selection and provided some reference to be implemented in breeding programs to improve the mastitis resistance of dairy cattle.


Subject(s)
Haplotypes , Milk/cytology , Polymorphism, Single Nucleotide , Toll-Like Receptor 4/genetics , Alleles , Animals , Breeding , Cattle , Female , Gene Frequency , Genotype , Mastitis, Bovine/genetics , Mastitis, Bovine/pathology
12.
PLoS One ; 8(6): e67637, 2013.
Article in English | MEDLINE | ID: mdl-23840757

ABSTRACT

The canonical transient receptor potential (TRPC) channels are Ca(2+)-permeable cationic channels controlling the Ca(2+) influx evoked by G protein-coupled receptor activation and/or by Ca(2+) store depletion. Here we investigate the involvement of TRPCs in the cell differentiation of lung cancer. The expression of TRPCs and the correlation to cancer differentiation grade in non-small cell lung cancer (NSCLC) were analyzed by real-time PCR and immunostaining using tissue microarrays from 28 patient lung cancer samples. The association of TRPCs with cell differentiation was also investigated in the lung cancer cell line A549 by PCR and Western blotting. The channel activity was monitored by Ca(2+) imaging and patch recording after treatment with all-trans-retinoic acid (ATRA). The expression of TRPC1, 3, 4 and 6 was correlated to the differentiation grade of NSCLC in patients, but there was no correlation to age, sex, smoking history and lung cancer cell type. ATRA upregulated TRPC3, TRPC4 and TRPC6 expression and enhanced Ca(2+) influx in A549 cells, however, ATRA showed no direct effect on TRPC channels. Inhibition of TRPC channels by pore-blocking antibodies decreased the cell mitosis, which was counteracted by chronic treatment with ATRA. Blockade of TRPC channels inhibited A549 cell proliferation, while overexpression of TRPCs increased the proliferation. We conclude that TRPC expression correlates to lung cancer differentiation. TRPCs mediate the pharmacological effect of ATRA and play important roles in regulating lung cancer cell differentiation and proliferation, which gives a new understanding of lung cancer biology and potential anti-cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Differentiation/genetics , Lung Neoplasms/pathology , Transient Receptor Potential Channels/genetics , Calcium/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Transient Receptor Potential Channels/metabolism , Tretinoin/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
13.
Yi Chuan ; 35(6): 771-7, 2013 Jun.
Article in Chinese | MEDLINE | ID: mdl-23774022

ABSTRACT

As a useful tool for genetic engineering, piggyBac (PB) transposons have been widely used in more than one species of transgenosis or generating mutation studies. At present, the studies about PB transposons in cattle were few. In order to get the PB transposon integration sites and summarize its characteristics in bovine genome, donor plasmid of PB[CMV-EGFP] and helper-dependent plasmid of pcDNA-PBase were constructed and transferred into bovine fibroblasts by Amaxa basic nucleofector kit for primary mammalian fibroblasts. Cell clones stably transfected were obtained after screening by G-418. Genomic DNA of transgenic cells was extracted and the integration sites of PB transposon were detected by genome walking technology. Eight integration sites were obtained in bovine genome, although only 5 sites were mapped on chromosomes 1, 2, 11, and X chromosome. We found that PB transposon was inserted into the "TTAA" location and integrated into the intergenic non-regulatory sites between two genes. Analysis of the composition of the five bases, which was close to the side of the PB integration sites "TTAA", showed that PB 5' tended to be inserted into region rich in GC (62.5%). From the study, we got that transposition occurred in cattle genome by PB transposons and the integration site information acquired from the research will provide theoretical references for bovine study by PB transposon.


Subject(s)
Cattle/genetics , DNA Transposable Elements , Animals , Genome , Plasmids , Transfection
14.
Curr Cancer Drug Targets ; 13(1): 103-16, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22920441

ABSTRACT

TRPC channels are Ca²âº-permeable cationic channels controlling Ca²âº influx response to the activation of G protein-coupled receptors and protein tyrosine kinase pathways or the depletion of Ca²âº stores. Here we aimed to investigate whether TRPC can act as the potential therapeutic targets for ovarian cancer. The mRNAs of TRPC1, TRPC3, TRPC4 and TRPC6 were detected in human ovarian adenocarcinoma. The spliced variants of TRPC1ß, TRPC3a, TRPC4ß, TRPC4γ, and TRPC6 with exon 3 and 4 deletion were highly expressed in the ovarian cancer cells, and a novel spliced isoform of TRPC1 with exon 9 deletion (TRPC1(E9del)) was identified. TRPC proteins were also detected by Western blotting and immunostaining. The expression of TRPC1, TRPC3, TRPC4 and TRPC6 was significantly lower in the undifferentiated ovarian cancer cells, but all-trans retinoic acid up-regulated the gene expression of TRPCs. The expression level was correlated to the cancer differentiation grade. The non-selective TRPC channel blockers, 2-APB and SKF-96365, significantly inhibited the cell proliferation, whilst the increase of TRPC channel activity by trypsin promoted the cell proliferation. Transfection with siRNA targeting TRPC1, TRPC3, TRPC4 and TRPC6 or application of specific blocking antibodies targeting to TRPC channels inhibited the cell proliferation. On the contrary, overexpression of TRPC1, TRPC1(E9del), TRPC3, TRPC4, and TRPC6 increased the cancer cell colony growth. These results suggest that TRPCs and their spliced variants are important for human ovarian cancer development and alteration of the expression or activity of these channels could be a new strategy for anticancer therapy.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , TRPC Cation Channels/metabolism , Adenocarcinoma/pathology , Alternative Splicing/drug effects , Amino Acid Sequence , Antibodies, Blocking , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Membrane Transport Modulators/pharmacology , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Ovarian Neoplasms/pathology , Ovary/drug effects , Ovary/pathology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics , Tretinoin/pharmacology
15.
Biochem Pharmacol ; 84(8): 1024-35, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22842488

ABSTRACT

STIM1 is a Ca(2+) sensing molecule. Once the Ca(2+) stores are depleted, STIM1 moves towards the plasma membrane (PM) (translocation), forms puncta (clustering), and triggers store-operated Ca(2+) entry (SOCE). Although this process has been regarded as a main mechanism for store-operated Ca(2+) channel activation, the STIM1 clustering is still unclear. Here we discovered a new phenomenon of STIM1 clustering, which is not triggered by endoplasmic reticulum (ER) Ca(2+) depletion. STIM1 subplasmalemmal translocation and clustering can be induced by ER Ca(2+) store depletion with thapsigargin (TG), G-protein-coupled receptor activator trypsin and ryanodine receptor (RyR) agonists caffeine and 4-chloro-3-ethylphenol (4-CEP) in the HEK293 cells stably transfected with STIM1-EYFP. The STIM1 clustering induced by TG was more sustained than that induced by trypsin and RyR agonists. Interestingly, 4-CEP-induced STIM1 clustering also happened in the cytosol without ER Ca(2+) store depletion. Application of some pharmacological regulators including flufenamic acid, 2-APB, and carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP) at concentrations without affecting ER Ca(2+) store also evoked cytosolic STIM1 clustering. However, the direct store-operated ORAI channel blockers (SKF-96365, Gd(3+) and diethylstilbestrol) or the signaling pathway inhibitors (genistein, wortmannin, Y-27632, forskolin and GF109203X) did not change the STIM1 movement. Disruption of cytoskeleton by colchicine and cytochalasin D also showed no effect on STIM1 movement. We concluded that STIM1 clustering and translocation are two dynamic processes that can be pharmacologically dissociated. The ER Ca(2+) store-independent mechanism for STIM1 clustering is a new alternative mechanism for regulating store-operated channel activity, which could act as a new pharmacological target.


Subject(s)
Calcium/metabolism , Cytosol/drug effects , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Ion Transport , Mitochondria/metabolism , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Stromal Interaction Molecule 1
16.
Biochem Biophys Res Commun ; 424(2): 279-84, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22750002

ABSTRACT

Transient receptor potential melastatin 2 (TRPM2) is a Ca(2+)-permeable cationic channel in the TRP channel family. The channel activity can be regulated by reactive oxygen species (ROS) and cellular acidification, which has been implicated to the pathogenesis of diabetes and some neuronal disorders. However, little is known about the effect of redox-active metal ions, such as copper, on TRPM2 channels. Here we investigated the effect of divalent copper on TRPM2. TRPM2 channel was over-expressed in HEK-293 cells and the whole-cell current was recorded by patch clamp. We found the whole-cell current evoked by intracellular ADP-ribose was potently inhibited by Cu(2+) with a half maximal inhibitory concentration (IC(50)) of 2.59 µM. The inhibitory effect was irreversible. The single channel activity was abolished in the outside-out patches, and intracellular application of Cu(2+) did not prevent the channel activation, suggesting that the action site of Cu(2+) is located in the extracellular domains of the channel. TRPM2 current was also blocked by Hg(2+), Pb(2+), Fe(2+) and Se(2+). We concluded that Cu(2+) is a potent TRPM2 channel blocker. The sensitivity of TRPM2 channel to heavy metal ions could be a new mechanism for the pathogenesis of some metal ion-related diseases.


Subject(s)
Cations, Divalent/pharmacology , Copper/pharmacology , TRPM Cation Channels/antagonists & inhibitors , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Cations, Divalent/metabolism , Copper/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Iron/metabolism , Iron/pharmacology , Lead/metabolism , Lead/pharmacology , Mercury/metabolism , Mercury/pharmacology , Patch-Clamp Techniques , Selenium/metabolism , Selenium/pharmacology
17.
Br J Pharmacol ; 167(6): 1232-43, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22646516

ABSTRACT

BACKGROUND AND PURPOSE: Fenamate analogues, econazole and 2-aminoethoxydiphenyl borate (2-APB) are inhibitors of transient receptor potential melastatin 2 (TRPM2) channels and are used as research tools. However, these compounds have different chemical structures and therapeutic applications. Here we have investigated the pharmacological profile of TRPM2 channels by application of newly synthesized fenamate analogues and the existing channel blockers. EXPERIMENTAL APPROACH: Human TRPM2 channels in tetracycline-regulated pcDNA4/TO vectors were transfected into HEK293 T-REx cells and the expression was induced by tetracycline. Whole cell currents were recorded by patch-clamp techniques. Ca(2+) influx or release was monitored by fluorometry. KEY RESULTS: Flufenamic acid (FFA), mefenamic acid (MFA) and niflumic acid (NFA) concentration-dependently inhibited TRPM2 current with potency order FFA > MFA = NFA. Modification of the 2-phenylamino ring by substitution of the trifluoromethyl group in FFA with -CH(3), -F, -CF(3), -OCH(3), -OCH(2)CH(3), -COOH, and -NO(2) at various positions, reduced channel blocking potency. The conservative substitution of 3-CF(3) in FFA by -CH(3) (3-MFA), however, gave the most potent fenamate analogue with an IC(50) of 76 µM, comparable to that of FFA, but unlike FFA, had no effect on Ca(2+) release. 3-MFA and FFA inhibited the channel intracellularly. Econazole and 2-APB showed non-selectivity by altering cytosolic Ca(2+) movement. Econazole also evoked a non-selective current. CONCLUSION AND IMPLICATIONS: The fenamate analogue 3-MFA was more selective than other TRPM2 channel blockers. FFA, 2-APB and econazole should be used with caution as TRPM2 channel blockers, as these compounds can interfere with intracellular Ca(2+) movement.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Boron Compounds/pharmacology , Econazole/pharmacology , Fenamates/pharmacology , TRPM Cation Channels/antagonists & inhibitors , Calcium/physiology , HEK293 Cells , Humans
18.
Biochem Pharmacol ; 83(7): 923-31, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22285229

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used anti-inflammatory therapeutic agents, among which the fenamate analogues play important roles in regulating intracellular Ca²âº transient and ion channels. However, the effect of NSAIDs on TRPC4 and TRPC5 is still unknown. To understand the structure-activity of fenamate analogues on TRPC channels, we have synthesized a series of fenamate analogues and investigated their effects on TRPC4 and TRPC5 channels. Human TRPC4 and TRPC5 cDNAs in tetracycline-regulated vectors were transfected into HEK293 T-REx cells. The whole cell current and Ca²âº movement were recorded by patch clamp and calcium imaging, respectively. Flufenamic acid (FFA), mefenamic acid (MFA), niflumic acid (NFA) and diclofenac sodium (DFS) showed inhibition on TRPC4 and TRPC5 channels in a concentration-dependent manner. The potency was FFA>MFA>NFA>DFS. Modification of 2-phenylamino ring by substitution of the trifluoromethyl group in FFA with F, CH3, OCH3, OCH2CH3, COOH, and NO2 led to the changes in their channel blocking activity. However, 2-(2'-methoxy-5'-methylphenyl)aminobenzoic acid stimulated TRPC4 and TRPC5 channels. Selective COX1-3 inhibitors (aspirin, celecoxib, acetaminophen, and indomethacin) had no effect on the channels. Longer perfusion (> 5 min) with FFA (100 µM) and MFA (100 µM) caused a potentiation of TRPC4 and TRPC5 currents after their initial blocking effects that appeared to be partially mediated by the mitochondrial Ca²âº release. Our results suggest that fenamate analogues are direct modulators of TRPC4 and TRPC5 channels. The substitution pattern and conformation of the 2-phenylamino ring could alter their blocking activity, which is important for understanding fenamate pharmacology and new drug development targeting the TRPC channels.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Calcium/metabolism , Fenamates/pharmacology , TRPC Cation Channels/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Culture Techniques , Fenamates/chemical synthesis , Fenamates/chemistry , Fluorescent Dyes/chemistry , Fura-2/analogs & derivatives , Fura-2/chemistry , HEK293 Cells , Humans , Male , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Muscle, Smooth, Vascular/cytology , Patch-Clamp Techniques , Rats , Structure-Activity Relationship , TRPC Cation Channels/genetics , Transfection
19.
Toxicol Sci ; 125(1): 56-68, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21984481

ABSTRACT

Mercury is an established worldwide environmental pollutant with well-known toxicity affecting neurodevelopment in humans, but the molecular basis of cytotoxicity and the detoxification procedure are still unclear. Here we examined the involvement of the canonical transient receptor potential (TRPC) channel in the mercury-induced cytotoxicity and the potential detoxification strategy. Whole-cell and excised patches, Ca(2+) imaging, and site-directed mutagenesis were used to determine the mechanism of action of mercurial compounds on TRPC channels overexpressed in HEK293 cells, and cytotoxicity and preventive effect were investigated in cell culture models using small interfering RNA and pharmacological blockers. Mercury potently activates TRPC4 and TRPC5 channels. The extracellular cysteine residues (C(553) and C(558)) near the channel pore region of TRPC5 are the molecular targets for channel activation by mercury. The sensitivity of mercury to TRPC5 is presumed to be specific because other divalent heavy metal pollutants, such as Cd(2+), Ni(2+), and Zn(2+), had no stimulating effect, and TRPC3, TRPC6, TRPV1, and TRPM2 were resistant to mercurial compounds. The channel activity of TRPC5, as well as TRPC4, induced by mercury, was prevented by 2-aminoethoxydiphenyl borate and modified by a reducing environment. The inhibition of TRPC5 channels by specific TRPC5 pore-blocking antibody or by SKF-96365 alleviated the cytotoxicity, whereas the mercury chelator, meso-2,3-dimercaptosuccinic acid, showed nonselective prevention of cell survival. Silencing of the TRPC5 gene reduced the mercury-induced neuronal damage. These results indicate that mercurial compounds are activators for TRPC5 and TRPC4 channels. Blockade of TRPC channels could be a novel strategy for preventing mercury-induced cytotoxicity and neurodevelopment impairment.


Subject(s)
Environmental Pollutants/toxicity , Mercuric Chloride/toxicity , Methylmercury Compounds/toxicity , TRPC Cation Channels/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , PC12 Cells , Protein Binding , Rats , Real-Time Polymerase Chain Reaction , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , Transfection
20.
Waste Manag Res ; 29(12): 1262-70, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21447611

ABSTRACT

Waste vinegar residue, the by-product of vinegar processing, was used as substrate for phytase production from Aspergillus ficuum NTG-23 in solid-state fermentation to investigate the potential for the efficient re-utilization or recycling of waste vinegar residue. Statistical designs were applied in the processing of phytase production. First, a Plackett-Burman (PB) design was used to evaluate eleven parameters: glucose, starch, wheat bran, (NH(4))(2)SO(4), NH(4)NO(3), tryptone, soybean meal, MgSO(4)·7H(2)O, CaCl(2)·7H(2)O, FeSO(4)·7H(2)O, incubation time. The PB experiments showed that there were three significant factors: glucose, soybean meal and incubation time. The closest values to the optimum point were then derived by steepest ascent path. Finally, a mathematical model was created and validated to explain the behavioural process after these three significant factors were optimized using response surface methodology (RSM). The best phytase activity was attained using the following conditions: glucose (7.2%), soybean meal (5.1%), and incubation time (271 h). The phytase activity was 7.34-fold higher due to optimization by PB design, steepest ascent path design and RSM. The phytase activity was enhanced 0.26-fold in comparison with the results by the second step of steepest ascent path design. The results indicate that with waste vinegar residue as a substrate higher production of phytase from Aspergillus ficuum NTG-23 could be obtained through an optimization process and that this method might be applied to an integrated system for recycling of the waste vinegar residue.


Subject(s)
6-Phytase/metabolism , Acetic Acid/chemistry , Aspergillus/metabolism , Waste Management/methods , Acetic Acid/metabolism , Models, Biological , Waste Products
SELECTION OF CITATIONS
SEARCH DETAIL
...