Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Pharmaceuticals (Basel) ; 17(3)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38543130

ABSTRACT

BACKGROUND: Transient receptor potential channels (TRP) are overexpressed in some pancreatic adenocarcinoma (PDAC) patients and cell lines, settling them as putative therapeutic targets in this disease. Reactive oxygen species (ROS), with levels increased in PDAC, modulate some members of the TRP family renamed "redox channels". Here, we investigate the direct effects of 4-hydroxinonenal (4-HNE) on TRPA1, natively expressed in PDAC cell lines and in association with cell migration and cell cycle progression. METHODS: We performed microfluorimetry experiments, while the activation of resident membrane channels was investigated using confocal microscopy. We applied a prospective molecular docking of 4-HNE using Autodock and AutoDock Tools4. Also, we simulated the diffusion of 4-HNE through the membrane from the extracellular space with the Permeability of Molecules across Membranes (PerMM) web server. The analysis of cell migration was performed using the wound healing assay, and cell cycle progression was acquired using a Beckman Coulter CytoFlex flow cytometer. RESULTS: Our results show, for the first time in PDAC, that 4-HNE diffuses through the cell membrane and rapidly activates Ca2+ uptake in PDAC cells. This process depends on TRPA1 activation, as 4-HNE forms a covalent binding with a pocket-like region within the intracellular N-terminal of the channel, shaped by the cysteine residues 621, 641, and 665. The activation of TRPA1 by 4-HNE inhibits cell migration and induces cell cycle arrest in the G2/M phase. CONCLUSIONS: Our study brings new insights into the effects of 4-HNE, highlighting the activation of the TRPA1 channel, a druggable, putative target for PDAC-expressing tumors.

2.
Antioxidants (Basel) ; 12(7)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37507867

ABSTRACT

The channels from the superfamily of transient receptor potential (TRP) activated by reactive oxygen species (ROS) can be defined as redox channels. Those with the best exposure of the cysteine residues and, hence, the most sensitive to oxidative stress are TRPC4, TRPC5, TRPV1, TRPV4, and TRPA1, while others, such as TRPC3, TRPM2, and TRPM7, are indirectly activated by ROS. Furthermore, activation by ROS has different effects on the tumorigenic process: some TRP channels may, upon activation, stimulate proliferation, apoptosis, or migration of cancer cells, while others inhibit these processes, depending on the cancer type, tumoral microenvironment, and, finally, on the methods used for evaluation. Therefore, using these polymodal proteins as therapeutic targets is still an unmet need, despite their draggability and modulation by simple and mostly unharmful compounds. This review intended to create some cellular models of the interaction between oxidative stress, TRP channels, and inflammation. Although somewhat crosstalk between the three actors was rather theoretical, we intended to gather the recently published data and proposed pathways of cancer inhibition using modulators of TRP proteins, hoping that the experimental data corroborated clinical information may finally bring the results from the bench to the bedside.

4.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012311

ABSTRACT

Pancreatic adenocarcinoma (PDAC) has low survival rates worldwide due to its tendency to be detected late and its characteristic desmoplastic reaction, which slows the use of targeted therapies. As such, the discovery of new connections between genes and the clinicopathological parameters contribute to the search for new biomarkers or targets for therapy. Transient receptor potential (TRP) channels are promising tools for cancer therapy or markers for PDAC. Therefore, in this study, we selected several genes encoding TRP proteins previously reported in cellular models, namely, Transient Receptor Potential Cation Channel Subfamily V Member 6 (TRPV6), Transient receptor potential ankyrin 1 (TRPA1), and Transient receptor potential cation channel subfamily M (melastatin) member 8 (TRPM8), as well as the TRPM8 Channel Associated Factor 1 (TCAF1) and TRPM8 Channel Associated Factor 2 (TCAF2) proteins, as regulatory factors. We analyzed the expression levels of tumors from patients enrolled in public datasets and confirmed the results with a validation cohort of PDAC patients enrolled in the Clinical Institute Fundeni, Romania. We found significantly higher expression levels of TRPA1, TRPM8, and TCAF1/F2 in tumoral tissues compared to normal tissues, but lower expression levels of TRPV6, suggesting that TRP channels have either tumor-suppressive or oncogenic roles. The expression levels were correlated with the tumoral stages and are related to the genes involved in calcium homeostasis (Calbindin 1 or S100A4) or to proteins participating in metastasis (PTPN1). We conclude that the selected TRP proteins provide new insights in the search for targets and biomarkers needed for therapeutic strategies for PDAC treatment.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , TRPM Cation Channels , Transient Receptor Potential Channels , Adenocarcinoma/pathology , Humans , Membrane Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Pancreatic Neoplasms
5.
Molecules ; 27(10)2022 May 19.
Article in English | MEDLINE | ID: mdl-35630748

ABSTRACT

THz spectroscopy is important for the study of ion channels because it directly addresses the low frequency collective motions relevant for their function. Here we used THz spectroscopy to investigate the inhibition of the epithelial sodium channel (ENaC) by its specific blocker, amiloride. Experiments were performed on A6 cells' suspensions, which are cells overexpressing ENaC derived from Xenopus laevis kidney. THz spectra were investigated with or without amiloride. When ENaC was inhibited by amiloride, a substantial increase in THz absorption was noticed. Molecular modeling methods were used to explain the observed spectroscopic differences. THz spectra were simulated using the structural models of ENaC and ENaC-amiloride complexes built here. The agreement between the experiment and the simulations allowed us to validate the structural models and to describe the amiloride dynamics inside the channel pore. The amiloride binding site validated using THz spectroscopy agrees with previous mutagenesis studies. Altogether, our results show that THz spectroscopy can be successfully used to discriminate between native and inhibited ENaC channels and to characterize the dynamics of channels in the presence of their specific antagonist.


Subject(s)
Amiloride , Epithelial Sodium Channels , Amiloride/metabolism , Amiloride/pharmacology , Animals , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Oocytes/metabolism , Spectrum Analysis , Xenopus laevis/metabolism
6.
Int J Mol Sci ; 22(9)2021 May 08.
Article in English | MEDLINE | ID: mdl-34066780

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide and the second most common cause of cancer-related death globally. This type of liver cancer is frequently detected at a late stage by current biomarkers because of the high clinical and biological heterogeneity of HCC tumours. From a plethora of molecules and cellular compounds, small nanoparticles with an endosomal origin are valuable cancer biomarkers or cargos for novel treatments. Despite their small sizes, in the range of 40-150 nm, these particles are delimited by a lipid bilayer membrane with a specific lipid composition and carry functional information-RNA, proteins, miRNAs, long non-coding RNAs (lncRNAs), or DNA fragments. This review summarizes the role of exosomal microRNA (miRNA) species as biomarkers in HCC therapy. After we briefly introduce the exosome biogenesis and the methods of isolation and characterization, we discuss miRNA's correlation with the diagnosis and prognosis of HCC, either as single miRNA species, or as specific panels with greater clinical impact. We also review the role of exosomal miRNAs in the tumourigenic process and in the cell communication pathways through the delivery of cargos, including proteins or specific drugs.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Exosomes/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , MicroRNAs/genetics , Molecular Targeted Therapy , Animals , Biomarkers, Tumor/metabolism , Disease Progression , Humans , MicroRNAs/metabolism
8.
Sci Rep ; 11(1): 2018, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479347

ABSTRACT

The transient receptor potential ankyrin type 1 (TRPA1) channel belongs to the TRP superfamily of ion channels. TRPA1 is a membrane protein with multiple functions able to respond to noxious stimuli, reactive oxygen species, inflammatory cytokines or pungent substances, and it participates in pain signalling, taste, inflammation and various steps of the tumorigenic process. To date, no reports have addressed the expression and function of TRPA1 in pancreatic ductal adenocarcinoma (PDAC) cells. This work reports the endogenous expression of TRPA1 channels in human pancreatic adenocarcinoma cell lines and provides insights into the function of the TRPA1 protein in the Panc-1 cell line. This study reports that cell lines isolated from PDAC patients had different levels of TRPA1 expression. The channel activity in Panc-1 cells, as assessed with electrophysiological (whole-cell patch clamp) and microfluorimetry methods, showed that non-selective cationic currents were activated by allyl isothiocyanate (AITC) in Panc-1 cells and inhibited by the selective TRPA1 antagonist A-967079. The current elicited by the specific agonist was associated with a robust increase in intracellular Ca2+. Furthermore, siRNA-induced downregulation of TRPA1 enhanced cell migration in the wound healing assay, indicating a possible role of ion channels independent from pore function. Finally, TRPA1 activation changed the cell cycle progression. Taken together, these results support the idea of channel-dependent and independent role for TRPA1 in tumoral processes.


Subject(s)
Adenocarcinoma/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , TRPA1 Cation Channel/genetics , Adenocarcinoma/pathology , Calcium/metabolism , Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Electrophysiological Phenomena , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Potentials/drug effects , Oximes/pharmacology , Patch-Clamp Techniques , TRPA1 Cation Channel/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
9.
Cancers (Basel) ; 12(5)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443546

ABSTRACT

Background: Improving surgical outcomes in hepatocellular carcinoma (HCC) patients would greatly benefit from biomarkers. Angiogenesis and inflammation are hallmarks of HCC progression and therapeutic targets. Methods: We retrospectively evaluated preoperative clinical variables and circulating (plasma) biomarkers of angiogenesis and inflammation in a cohort of HCC patients who underwent liver resection (LR) or transplantation (LT). Biomarker correlation with outcomes-freedom of liver recurrence (FLR), disease-free survival (DFS) and overall survival (OS)-was tested using univariate and multivariate Cox regression analyses. Results: Survival outcomes associated with sVEGFR1, VEGF and VEGF-C in LT patients and with IL-10 in LR patients. Moreover, in LT patients within Milan criteria, higher plasma VEGF and sVEGFR1 were associated with worse outcomes, while in those outside Milan criteria lower plasma VEGF-C associated with better outcomes. Multivariate analysis indicated that adding plasma VEGF or VEGF-C to a predictive model including Milan criteria and AFP improved prediction of DFS and OS (all p < 0.05). Conclusion: Survival outcomes after LR or LT differentially associated with angiogenic and inflammatory biomarkers. High plasma VEGF correlated with poorer prognosis within Milan criteria while low plasma VEGF-C associated with better prognosis outside Milan criteria. These candidate biomarkers should be further validated to improve patient stratification.

10.
Biochim Biophys Acta Gen Subj ; 1864(7): 129580, 2020 07.
Article in English | MEDLINE | ID: mdl-32109505

ABSTRACT

TRPM8 member of the TRP superfamily of membrane proteins participates to various cellular processes ranging from Ca2+ uptake and cold sensation to cellular proliferation and migration. TRPM8 is a large tetrameric protein with more than 70% of its residues located in the cytoplasm. TRPM8 is N-glycosylated, with a single site per subunit. This work focuses on the N-glycosylation of TRPM8 channel that was previously studied by our group in relation to proliferation and migration of tumoral cells. Here, experimental data performed with deglycosylating agents assess that the sole glycosylation site contains complex glycans with a molecular weight of 2.5 kDa. The glycosylation state of TRPM8 in cells untreated and treated with a deglycosylating agent was addressed with Terahertz (THz) spectroscopy. Results show a clear difference between cells comprising glycosylated and deglycosylated TRPM8, the first presenting an increased THz absorption. Human TRPM8 was modelled using as templates the available TRPM8 and other TRPM channels structures. Glycosylations were modelled by considering two glycan structures with molecular weight close to the experiment: shorter and branched at the first sugar unit (glc1) and longer and unbranched (glc2). Simulation of THz spectra based on the molecular dynamics of unglycosylated and the two glycosylated TRPM8 models in lipid membrane and solvation box showed that glycan structure strongly influences the THz spectrum of the channel and of other components from the simulation system. Only spectra of TRPM8 with glc1 glycans were in agreement with the experiment, leading to the validation of glc1 glycan structure.


Subject(s)
Glucans/chemistry , Membrane Lipids/chemistry , Models, Molecular , TRPM Cation Channels/chemistry , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Glycosylation , Humans , Sugars/chemistry , TRPM Cation Channels/ultrastructure , Terahertz Spectroscopy
11.
BMC Cancer ; 19(1): 1007, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31660891

ABSTRACT

BACKGROUND: The expression of microRNAs (miRNAs) is a promising prognostic and diagnostic tool in hepatocellular carcinoma (HCC). Here we performed small RNA sequencing (sRNA-seq) of tissue, serum and serum exosomes to investigate changes in miRNA expression between the different sample types and correlated the expression with clinical parameters. We also performed gene expression arrays on tumor and normal tissue. RESULTS: Paired tissue, serum and serum exosomes sequencing revealed consistent positive correlation of miR-21 between serum exosomes and tumor tissue, indicating that miR-21 could be exported from tissue to circulation via exosomes. We found that let-7 miRNAs are generally upregulated in serum exosomes compared to whole serum, indicating that these miRNAs could be preferentially loaded into exosomes. Comparing serum from HCC patients with serum from healthy individuals revealed a global increase of miRNAs in serum from HCC patients, including an almost 4-fold increase of several miRNAs, including the liver-specific miR-122. When correlating miRNA expression with clinical parameters we detected significant association between hepatitis B virus (HBV) infection and miR-122 in serum as well as several serum and tissue-miRNAs that correlated with surgery type. We found that miR-141 and miR-146 correlated with cirrhosis in tumor tissue and normal tissue, respectively. Finally, high expression of miR-21 in tumors were associated with poor survival. Focusing on gene expression we found several significant messenger RNAs (mRNAs) between tumor and normal tissue and a Gene Ontology (GO) analysis revealed that these changes were mainly related to cell cycle and metabolism. Further, we detected mRNAs that correlated with cirrhosis and HBV infection in tissue. Finally, GO analysis of predicted targets for miRNAs down-regulated in tumor found that these were enriched for functions related to collagen synthesis. CONCLUSIONS: Our combined data point to altered miRNA and mRNA expression contributing to both generally impaired lipid metabolism and increased cell proliferation and a miRNA-driven increase in collagen synthesis in HCC. Our results further indicate a correlation in miRNA expression between exosomes, serum, and tissue samples suggesting export from tumors via exosomes. This correlation could provide a basis for a more tumor-specific miRNA profile in serum.


Subject(s)
Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Exosomes/genetics , Liver Neoplasms/blood , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Messenger/genetics , Transcriptome/genetics , Biomarkers, Tumor/genetics , Cohort Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hepatitis B/genetics , Hepatitis B/virology , Hepatitis B virus , Humans , Liver Cirrhosis/genetics , Prognosis , RNA-Seq/methods
12.
Tumour Biol ; 39(8): 1010428317720940, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28857015

ABSTRACT

Transient receptor potential melastatin 8 (TRPM8), a membrane ion channel, is activated by thermal and chemical stimuli. In pancreatic ductal adenocarcinoma, TRPM8 is required for cell migration, proliferation, and senescence and is associated with tumor size and pancreatic ductal adenocarcinoma stages. Although the underlying mechanisms of these processes have yet to be described, this cation-permeable channel has been proposed as an oncological target. In this study, the glycosylation status of the TRPM8 channel was shown to affect cell proliferation, cell migration, and calcium uptake. TRPM8 expressed in the membrane of the Panc-1 pancreatic tumoral cell line is non-glycosylated, whereas human embryonic kidney cells transfected with human TRPM8 overexpress a glycosylated protein. Moreover, our data suggest that Ca2+ uptake is modulated by the glycosylation status of the protein, thus affecting cell proliferation.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , TRPM Cation Channels/genetics , Calcium/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Glycosylation , Humans , Pancreas , Pancreatic Neoplasms/pathology , Patch-Clamp Techniques
13.
Gen Physiol Biophys ; 35(3): 259-71, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27045669

ABSTRACT

The function of the epithelial Na+ channel from the apical membrane of many Na+ transporting epithelia is modulated by various chemical compounds from the extracellular space, such as heavy metals, protons or chloride ions. We have studied the effect of extracellular Cd2+ on the function of the epithelial Na+ channel (ENaC) in heterologously expressed Xenopus laevis oocytes and Na+-transporting epithelia. We assayed channel function as the amiloride-sensitive sodium current (I(Na)). Cd2+ rapidly and voltage-independently inhibited INa in oocytes expressing αßγ Xenopus ENaC (xENaC). The extracellular Cd2+ inhibited Na+ transport and showed no influence on ENaC trafficking, as revealed by concomitant measurements of the transepithelial current, conductance and capacitance in Na+-transporting epithelia. Instead, amiloride inhibition was noticeably diminished in the presence of Cd2+ on the apical membrane. Using molecular modeling approaches, we describe the amiloride binding sites in rat and xENaC structures, and we present four putative binding sites for Cd2+. These results indicate that ENaC functions as a sensor for external Cd2+.


Subject(s)
Cadmium/administration & dosage , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/physiology , Oocytes/physiology , Sodium/metabolism , Animals , Binding Sites , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Sodium Channels/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Models, Chemical , Molecular Docking Simulation , Oocytes/drug effects , Protein Binding , Xenopus laevis
14.
Pancreas ; 43(5): 795-800, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24658318

ABSTRACT

OBJECTIVE: Recently, the transient receptor potential melastatin 8 (TRPM8) channel has emerged as a putative biomarker for pancreatic ductal adenocarcinoma (PDA). This study aimed to evaluate the expression of TRPM8 and its modulation by specific agonists and antagonists in PDA cells. METHODS: We examined the protein expression of TRPM8 in 3 different PDA cell lines and compared it with a nontumoral epithelial cell line of human pancreatic origin using Western blotting and immunocytochemical analysis. To assess the function of TRPM8 channels, we measured the TRPM8 currents in whole-cell mode of the patch clamp technique. To explore the putative involvement of TRPM8 in cell migration, we investigated the motility of PDA cells using the scratch-wound assay. RESULTS: Pancreatic ductal adenocarcinoma cells express functional plasma membrane TRPM8 channels, which are responsive after exposure to agonists (menthol and icilin) and antagonists N-(3-aminopropyl)-2-{[(3-methylphenyl) methyl]oxy}-N-(2-thienylmethyl)benzamide hydrochloride salt. The silencing of TRPM8 expression by small interfering RNA augments the migration of PDA cells. Conversely, the activated form of TRPM8 inhibits PDA cell motility. CONCLUSIONS: An unglycosylated TRPM8 protein is expressed and is functional in the membrane of PDA cells. Transient receptor potential melastatin 8 inhibits the migration of PDA cells, suggesting a putative role as a biomarker or target for this channel for PDA therapy.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , TRPM Cation Channels/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Benzamides/pharmacology , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/physiology , Cell Movement/drug effects , Cell Movement/genetics , HEK293 Cells , Humans , Immunohistochemistry , Membrane Potentials/drug effects , Membrane Proteins/genetics , Menthol/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Patch-Clamp Techniques , Pyrimidinones/pharmacology , RNA Interference , TRPM Cation Channels/agonists , TRPM Cation Channels/antagonists & inhibitors , Thiophenes/pharmacology , Pancreatic Neoplasms
15.
Nephron Extra ; 1(1): 24-37, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22470376

ABSTRACT

BACKGROUND/AIMS: The present study was conducted to investigate the renal tubular handling of inorganic cadmium (Cd(2+)) by exposing primary human tubular cell cultures to physiologically relevant doses of cadmium chloride (CdCl(2)). Furthermore, the cellular accumulation of Cd(2+) was compared to that of metallothionein-1-bound Cd (Cd7MT-1). Finally, this study aimed to investigate the effect of the accumulation of Cd (both Cd(2+) and Cd7MT-1) in renal cells on the expression of genes relevant to nephrotoxic processes. METHODS: Cd concentration was measured using atomic absorption spectrometry. mRNA expression was evaluated by quantitative real-time RT-PCR. RESULTS: Cd(2+) accumulated into human tubular cells in a concentration- and time-dependent way. Furthermore, cellular accumulation of Cd(2+) was different from the cellular accumulation of Cd7MT-1, indicative for different uptake routes. Finally, mRNA expression of the genes encoding the anti-oxidative proteins metallothionein-1 (MT-1) and heme-oxygenase-1 (HO-1) as well as the pro-apoptotic Bcl-2-associated X protein (Bax) were upregulated by CdCl(2) and not by Cd7MT1. CONCLUSION: In the presence of physiologically relevant Cd concentrations, tubular accumulation of the element in its inorganic form is different from that of Cd7MT-1. Furthermore, the tubular accumulation of inorganic Cd induces mRNA expression of genes of which the protein products may play a role in Cd-associated renal toxicity.

16.
Biosens Bioelectron ; 25(7): 1729-34, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20047824

ABSTRACT

Calcium oxalate is one of the main constituents of kidney stones and has a proved deleterious effect on renal cells that is mediated by oxidative stress. However, the subcellular source of this oxidative stress, and whether it is extending to the extracellular space or not, is still disputed. Therefore, an electrochemical superoxide biosensor was constructed, positioned above A6 renal cells, and used to measure in real-time the extracellular oxidative burst following addition of calcium oxalate crystals. It was observed that A6 cells do secrete superoxide into their extracellular space in few minutes after encountering calcium oxalate crystals. The amount of released superoxide peaks at about 20 min. Superoxide is cleared away from the extracellular space after approximately 3h. Superoxide secretion depends on the presence of superoxide-scavenging enzyme superoxide dismutase, the age of the cells, the amount of calcium oxalate crystals, and the temperature. Moreover, the effect of calcium oxalate crystals was mimicked by phorbol 12-myristate 13-acetate. The developed sensing system can be a useful tool for biologists investigating nephrolithiasis at cellular level.


Subject(s)
Biological Assay/instrumentation , Biosensing Techniques/instrumentation , Calcium Oxalate/administration & dosage , Kidney/drug effects , Kidney/metabolism , Oximetry/instrumentation , Respiratory Burst/physiology , Superoxides/metabolism , Biosensing Techniques/methods , Cells, Cultured , Computer Systems , Equipment Design , Equipment Failure Analysis , Humans , Kidney/cytology , Reproducibility of Results , Sensitivity and Specificity
17.
Am J Physiol Cell Physiol ; 289(4): C946-58, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15944207

ABSTRACT

The epithelial Na+ channel (ENaC) is modulated by various extracellular factors, including Na+, organic or inorganic cations, and serine proteases. To identify the effect of the divalent Ni2+ cation on ENaCs, we compared the Na+ permeability and amiloride kinetics of Xenopus ENaCs (xENaCs) and rat ENaCs (rENaCs) heterologously expressed in Xenopus oocytes. We found that the channel cloned from the kidney of the clawed toad Xenopus laevis [wild-type (WT) xENaC] was stimulated by external Ni2+, whereas the divalent cation inhibited the channel cloned from the rat colon (WT rENaC). The kinetics of amiloride binding were determined using noise analysis of blocker-induced fluctuation in current adapted for the transoocyte voltage-clamp method, and Na+ conductance was assessed using the dual electrode voltage-clamp (TEVC) technique. The inhibitory effect of Ni2+ on amiloride binding is not species dependent, because Ni2+ decreased the affinity (mainly reducing the association rate constant) of the blocker in both species in competition with Na+. Importantly, using the TEVC method, we found a prominent difference in channel conductance at hyperpolarizing voltage pulses. In WT xENaCs, the initial ohmic current response was stimulated by Ni2+, whereas the secondary voltage-activated current component remained unaffected. In WT rENaCs, only a voltage-dependent block by Ni2+ was obtained. To further study the origin of the xENaC stimulation by Ni2+, and based on the rationale of the well-known high affinity of Ni2+ for histidine residues, we designed alpha-subunit mutants of xENaCs by substituting histidines that were expressed in oocytes, together with WT beta- and gamma-subunits. Changing His215 to Asp in one putative amiloride-binding domain (WYRFHY) in the extracellular loop between Na+ channel membrane segments M1 and M2 had no influence on the stimulatory effect of Ni2+, and neither did complete deletion of this segment. Next, we mutated His416 flanked by His411 and Cys417, a unique site for possible heavy metal ion chelation, and, with this quality, most proximal (approximately 100 amino acids upstream of the second putative amiloride binding site at the pore entrance), was found localized at M2. Replacing His416 with arginine, aspartate, tyrosine, and alanine clearly affected amiloride binding in all cases, as well as Na+ conductance, as expressed in the xENaC current-voltage relationship, especially with regard to aspartate and tyrosine. However, similarly to those obtained with the WYRFHY stretch, none of these mutations could either abolish the stimulating effect of Ni2+ or reverse it to an inhibitory type.


Subject(s)
Nickel/pharmacology , Sodium Channels/drug effects , Amiloride/pharmacology , Animals , Diuretics/pharmacology , Electrophysiology , Epithelial Sodium Channels , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutation , Oocytes/metabolism , RNA, Messenger/metabolism , Rats , Recombinant Proteins/biosynthesis , Sodium Channels/biosynthesis , Sodium Channels/genetics , Sodium Channels/physiology , Species Specificity , Xenopus
18.
Pflugers Arch ; 447(6): 934-42, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14716490

ABSTRACT

We developed a non-invasive technique for electrophysiological investigations of ion transport proteins endogenously or heterologously expressed in Xenopus laevis oocytes. We named this technique the transoocyte voltage clamp (TOVC). Whereas in the classical two-microelectrode voltage-clamp (TEVC) technique, the oocyte is impaled with two glass microelectrodes, we mount the egg in a modified Ussing chamber as used for transepithelial electrophysiological studies. The oocyte is introduced in a container that is positioned between the two chamber halves. Proper fixation of the oocyte in the aperture of the container is accomplished under a stereo binocular microscope and the electrical seal between the oocyte and the container is achieved with silicon grease. The new method allows measurement of transoocyte currents and conductances as well as the recording of membrane impedance and the fluctuation analysis of ion currents. We studied a K+ channel that resembles the inward rectifier K+ channel endogenously expressed in Xenopus laevis oocytes. K+ currents were obtained by exposing one side of the oocyte to K(+)-containing solutions and by the application of different voltages. Adding Cs+ and Ba2+ inhibited these currents. The analysis of the fluctuation in current demonstrated a Lorentzian component in the power density spectrum. With the transoocyte voltage clamped to zero, the corner frequency (fc) was 61+/-1.7 Hz. Imposed positive transoocyte potentials caused a downward shift of fc. These findings are consistent with previous data obtained using the TEVC technique, and extend the characterization of the channel with kinetic data obtained from noise analysis.


Subject(s)
Oocytes/physiology , Patch-Clamp Techniques/methods , Animals , Electrophysiology , Female , In Vitro Techniques , Ion Channel Gating/physiology , Potassium Channels/physiology , Xenopus laevis
19.
FEBS Lett ; 515(1-3): 177-83, 2002 Mar 27.
Article in English | MEDLINE | ID: mdl-11943217

ABSTRACT

We used oocytes of the South African clawed toad Xenopus laevis to express the three subunits of the epithelial Na(+) channel from rat distal colon (rENaC). We combined conventional dual-microelectrode voltage-clamp with continuous capacitance (C(m)) measurements and noise analysis to evaluate the effects of cAMP and Ni(2+) on rENaC. Control oocytes or rENaC-expressing oocytes exhibited no spontaneous fluctuations in current. However, in rENaC-expressing oocytes amiloride induced a marked plateau-shaped rise of the power density spectra. Recordings using four different concentrations of amiloride revealed that the blocker-channel interactions were of the first order. A cocktail of the membrane permeant cAMP analogue chlorophenylthio-cAMP and IBMX (cAMP cocktail) increased amiloride-sensitive current (I(ami)) and conductance (G(ami)). Furthermore, C(m) was also increased following cAMP application, indicating an increase in plasma membrane surface area. Noise analysis showed that cAMP increased the number of active channels in the oocyte membrane while single-channel current decreased. From these data we conclude that cAMP triggered exocytotic delivery of preformed rENaCs to the plasma membrane. Ni(2+) (2.5 mM) inhibited about 60% of the rENaC current and conductance while C(m) remained unaffected. Noise analysis revealed that this inhibition could be attributed to a decrease in the apparent channel density, while single-channel current did not change significantly. These observations argue for direct effects of Ni(2+) on channel activity rather than induction of endocytotic removal of active channels from the plasma membrane.


Subject(s)
Cyclic AMP/pharmacology , Nickel/pharmacology , Oocytes/metabolism , Sodium Channels/biosynthesis , Sodium Channels/drug effects , Amiloride/pharmacology , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Electric Capacitance , Enzyme Inhibitors/pharmacology , Epithelial Sodium Channels , Exocytosis/drug effects , Intracellular Fluid/metabolism , Microinjections , Oocytes/cytology , Oocytes/drug effects , Patch-Clamp Techniques , Rats , Signal Processing, Computer-Assisted , Sodium/metabolism , Sodium Channels/genetics , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL