Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Pharmacol Rev ; 64(1): 1-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22090471

ABSTRACT

Calcium-activated chloride channels (CaCCs) are widely expressed in various tissues and implicated in physiological processes such as sensory transduction, epithelial secretion, and smooth muscle contraction. Transmembrane proteins with unknown function 16 (TMEM16A) has recently been identified as a major component of CaCCs. Detailed molecular analysis of TMEM16A will be needed to understand its structure-function relationships. The role this channel plays in physiological systems remains to be established and is currently a subject of intense investigation.


Subject(s)
Chloride Channels/physiology , Neoplasm Proteins/physiology , Xenopus Proteins/physiology , Animals , Anoctamin-1 , Chloride Channels/antagonists & inhibitors , Chloride Channels/biosynthesis , Chloride Channels/classification , Electrophysiological Phenomena , Glycosylation , Humans , International Agencies , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/classification , Organ Specificity , Phosphorylation , Protein Conformation , Terminology as Topic , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/biosynthesis , Xenopus Proteins/classification
2.
Annu Rev Physiol ; 72: 95-121, 2010.
Article in English | MEDLINE | ID: mdl-19827947

ABSTRACT

Until recently, anion (Cl(-)) channels have received considerably less attention than cation channels. One reason for this may be that many Cl(-) channels perform functions that might be considered cell-biological, like fluid secretion and cell volume regulation, whereas cation channels have historically been associated with cellular excitability, which typically happens more rapidly. In this review, we discuss the recent explosion of interest in Cl(-) channels, with special emphasis on new and often surprising developments over the past five years. This is exemplified by the findings that more than half of the ClC family members are antiporters, and not channels, as was previously thought, and that bestrophins, previously prime candidates for Ca(2+)-activated Cl(-) channels, have been supplanted by the newly discovered anoctamins and now hold a tenuous position in the Cl(-) channel world.


Subject(s)
Channelopathies/physiopathology , Chloride Channels/physiology , Amino Acid Sequence , Carrier Proteins/metabolism , Chloride Channels/classification , Chloride Channels/drug effects , Chloride Channels/genetics , History, 20th Century , Humans , Neoplasms/genetics , Neoplasms/pathology , Physiology/history
3.
Biophys J ; 98(10): 2163-9, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20483324

ABSTRACT

Several prokaryotic ClC proteins have been demonstrated to function as exchangers that transport both chloride ions and protons simultaneously in opposite directions. However, the path of the proton through the ClC exchanger, and how the protein brings about the coupled movement of both ions are still unknown. In this work, we use an atomistic molecular dynamics (MD) simulation to demonstrate that a previously unknown secondary water pore is formed inside an Escherichia coli ClC exchanger. The secondary water pore is bifurcated from the chloride ion pathway at E148. From the systematic simulations, we determined that the glutamate residue exposed to the intracellular solution, E203, plays an important role as a trigger for the formation of the secondary water pore, and that the highly conserved tyrosine residue Y445 functions as a barrier that separates the proton from the chloride ion pathways. Based on our simulation results, we conclude that protons in the ClC exchanger are conducted via a water network through the secondary water pore, and we propose a new mechanism for the coupled transport of chloride ions and protons. It has been reported that several members of ClC proteins are not just channels that simply transport chloride ions across lipid bilayers; rather, they are exchangers that transport both the chloride ion and proton in opposite directions. However, the ion transit pathways and the mechanism of the coupled movement of these two ions have not yet been unveiled. In this article, we report a new finding (to our knowledge) of a water pore inside a prokaryotic ClC protein as revealed by computer simulation. This water pore is bifurcated from the putative chloride ion, and water molecules inside the new pore connect two glutamate residues that are known to be key residues for proton transport. On the basis of our simulation results, we conclude that the water wire that is formed inside the newly found pore acts as a proton pathway, which enables us to resolve many problems that could not be addressed by previous experimental studies.


Subject(s)
Biological Transport/physiology , Chloride Channels/physiology , Chlorides/metabolism , Ion Transport/physiology , Molecular Dynamics Simulation/trends , Protons , Chloride Channels/classification , Chloride Channels/metabolism , Computer Simulation , Electric Conductivity , Escherichia coli/metabolism , Escherichia coli Proteins , Ion Channel Gating , Lipid Bilayers/pharmacology , Models, Molecular , Static Electricity , Structure-Activity Relationship , Water/physiology
4.
Exp Eye Res ; 90(6): 771-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20346358

ABSTRACT

Transport of water and electrolytes is critical for corneal clarity. Recent studies indicate another important function of transport of ions and electrolytes - establishing wound electric fields that guide cell migration. We found chloride (Cl(-)) flux is a major component of the corneal wound electric current. In order to elucidate the mechanisms of Cl(-) transport, we studied Cl(-) channels and transporters in human corneal epithelial (HCE) cells. We tested a transformed human corneal epithelial cell line (tHCE), primary cultures of human corneal epithelial cells (pHCE), and human donor corneas. We first used RT-PCR to determine expression levels of mRNA of CLC (Cl(-) channels/transporters of CLC gene family) family members and CFTR (cystic fibrosis transmembrane conductance regulator) in HCE cells. We then confirmed protein expression and distribution of selected CLC family members and CFTR with Western blot and immunofluorescence confocal microscopy. Finally, Cl(-) currents were recorded with electrophysiological techniques. The mRNAs of CLC-2, CLC-3, CLC-4, CLC-5, CLC-6, and CFTR were detected in the HCE cell line. CLC-1 and CLC-7 were not detectable. Western blot and immunostaining confirmed protein expression and distribution of CLC-2, CLC-3, CLC-4, CLC-6 and CFTR in human corneal epithelium. CLC-2 preferentially labeled the apical and basal layers, while CLC-3 and CLC-4 labeled only the superficial layer. CLC-6 and CFTR labeling showed a unique gradient with strong staining in apical layers which gradually decreased towards the basal layers. Corneal endothelium was positive for CLC-2, CLC-3, CLC-4, CLC-6 and possibly CFTR. Human corneal epithelial cells demonstrated voltage dependent Cl(-) currents. HCE cells express functional Cl(-) channels and transporters. CLC-2, CLC-3, CLC-4, CLC-6, and CFTR had distinct expression patterns in human corneal epithelium. Those molecules and their distribution may play important roles in maintaining resting Cl(-) fluxes and in regulating Cl(-) flux at corneal wounds, which may be a major contributor to wound electrical signaling.


Subject(s)
Anion Transport Proteins/genetics , Chloride Channels/genetics , Chlorides/metabolism , Epithelium, Corneal/metabolism , Gene Expression Regulation/physiology , Anion Transport Proteins/metabolism , Blotting, Western , Cell Line, Transformed , Chloride Channels/classification , Chloride Channels/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Electrophysiology , Endothelium, Corneal/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Ion Transport/physiology , Microscopy, Confocal , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Voltage-Dependent Anion Channels
5.
Biophys J ; 97(12): 3047-53, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20006941

ABSTRACT

Cl(-) channels play important roles in many physiological processes, including transepithelial ion absorption and secretion, smooth and skeletal muscle contraction, neuronal excitability, sensory perception, and cell volume regulation. The molecular identity of many types of Cl(-) channels is still unknown. Recently, three research groups have arrived independently at the identification of TMEM16A (also known as anoctamin-1) as a membrane protein strongly related to the activity of Ca(2+)-activated Cl(-) channels (CaCCs). Site-specific mutagenesis of TMEM16A alters the properties of the channels, thus suggesting that TMEM16A forms, at least in part, the CaCC. TMEM16A is a member of a family that includes nine other membrane proteins. All TMEM16 proteins have a similar structure, with eight putative transmembrane domains and cytosolic amino- and carboxy-termini. TMEM16B expression also evokes the appearance of CaCCs, but with biophysical characteristics (voltage dependence, unitary conductance) different from those associated to TMEM16A. The roles of the other TMEM16 proteins are still unknown. The study of TMEM16 proteins may lead to identification of novel molecular mechanisms underlying ion transport and channel gating by voltage and Ca(2+).


Subject(s)
Chloride Channels , Amino Acid Sequence , Animals , Chloride Channels/chemistry , Chloride Channels/classification , Chloride Channels/metabolism , Chlorine/metabolism , Electric Conductivity , Humans , Molecular Sequence Data
6.
BMC Dev Biol ; 9: 10, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19210762

ABSTRACT

BACKGROUND: Members of the calcium-activated chloride channel (CLCA) gene family have been suggested to possess a variety of functions including cell adhesion and tumor suppression. Expression of CLCA family members has mostly been analyzed in non-neural tissues. Here we describe the expression of mouse and human CLCA genes in the nervous system. RESULTS: We show that from the six mouse CLCA family members only Clca1, Clca2 and Clca4 mRNAs are expressed in the adult brain, predominantly in olfactory ensheathing cells. During mouse nervous system development Clca1/2 is more widely expressed, particularly in cranial nerves, the diencephalon and in the cerebral cortex. While human CLCA2 and CLCA4 genes are widely expressed in brain, and at particularly high levels in the optic nerve, human CLCA3, the closest homologue of mouse Clca1, Clca2 and Clca4, is not expressed in the brain. Furthermore, we characterize the expression pattern of mouse Clca1/2 genes during embryonic development by in situ hybridization. CONCLUSION: The data published in this article indicate that within the nervous system mouse Clca1/2 genes are highly expressed in the cells ensheathing cranial nerves. Human CLCA2 and CLCA4 mRNAs are expressed at high level in optic nerve. High level expression of CLCA family members in mouse and human glial cells ensheathing nerves suggests a specific role for CLCA proteins in the development and homeostasis of these cells.


Subject(s)
Chloride Channels/genetics , Multigene Family , Nervous System/metabolism , Adult , Animals , Base Sequence , Chloride Channels/classification , DNA Primers/genetics , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Mice , Nervous System/embryology , Nervous System/growth & development , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
7.
Science ; 364(6438): 395-399, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31023925

ABSTRACT

Severe local acidosis causes tissue damage and pain, and is one of the hallmarks of many diseases including ischemia, cancer, and inflammation. However, the molecular mechanisms of the cellular response to acid are not fully understood. We performed an unbiased RNA interference screen and identified PAC (TMEM206) as being essential for the widely observed proton-activated Cl- (PAC) currents (I Cl,H). Overexpression of human PAC in PAC knockout cells generated I Cl,H with the same characteristics as the endogenous ones. Zebrafish PAC encodes a PAC channel with distinct properties. Knockout of mouse Pac abolished I Cl,H in neurons and attenuated brain damage after ischemic stroke. The wide expression of PAC suggests a broad role for this conserved Cl- channel family in physiological and pathological processes associated with acidic pH.


Subject(s)
Chloride Channels/metabolism , Membrane Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Calcium/metabolism , Cell Death , Chloride Channels/classification , Chloride Channels/genetics , Chlorides/metabolism , Conserved Sequence , Evolution, Molecular , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Membrane Proteins/classification , Membrane Proteins/genetics , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Phylogeny , RNA Interference , Stroke/metabolism , Stroke/pathology , Zebrafish , Zebrafish Proteins/classification , Zebrafish Proteins/genetics
8.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 25(4): 980-3, 2008 Aug.
Article in Zh | MEDLINE | ID: mdl-18788323

ABSTRACT

It has been shown that a lot of diseases were related with the change or loss of Cl- channel functions. Among the Cl- channels, volume-regulated anion channel (VRAC) plays important roles in myocardial ischemia/reperfusion injury, cardiac arrhythmia and apoptosis; it may become a new target in the clinical treatment of heart diseases. This paper presents an overview of the physiological characteristics of VRAC and its relations with myocardial ischemia/reperfusion injury.


Subject(s)
Chloride Channels/physiology , Myocardial Reperfusion Injury/metabolism , Chloride Channels/classification , Chloride Channels/metabolism , Humans
9.
PLoS One ; 13(1): e0191512, 2018.
Article in English | MEDLINE | ID: mdl-29346439

ABSTRACT

Members of the chloride channel regulators, calcium-activated (CLCA) family, have been implicated in diverse biomedical conditions, including chronic inflammatory airway diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis, the activation of macrophages, and the growth and metastatic spread of tumor cells. Several observations, however, could not be repeated across species boundaries and increasing evidence suggests that select CLCA genes are particularly prone to dynamic species-specific evolvements. Here, we systematically characterized structural and expressional differences of the CLCA3 gene across mammalian species, revealing a spectrum of gene duplications, e.g., in mice and cows, and of gene silencing via diverse chromosomal modifications in pigs and many primates, including humans. In contrast, expression of a canonical CLCA3 protein from a single functional gene seems to be evolutionarily retained in carnivores, rabbits, guinea pigs, and horses. As an accepted asthma model, we chose the cat to establish the tissue and cellular expression pattern of the CLCA3 protein which was primarily found in mucin-producing cells of the respiratory tract and in stratified epithelia of the esophagus. Our results suggest that, among developmental differences in other CLCA genes, the CLCA3 gene possesses a particularly high dynamic evolutionary diversity with pivotal consequences for humans and other primates that seem to lack a CLCA3 protein. Our data also help to explain previous contradictory results on CLCA3 obtained from different species and warrant caution in extrapolating data from animal models in conditions where CLCA3 may be involved.


Subject(s)
Chloride Channels/physiology , Animals , Chloride Channels/classification , Evolution, Molecular , Multigene Family , Phylogeny , Respiratory Tract Diseases/genetics , Species Specificity
10.
Curr Biol ; 11(15): R620-8, 2001 Aug 07.
Article in English | MEDLINE | ID: mdl-11516971

ABSTRACT

Chloride (Cl(-)) is the most abundant extracellular anion in multicellular organisms. Passive movement of Cl(-) through membrane ion channels enables several cellular and physiological processes including transepithelial salt transport, electrical excitability, cell volume regulation and acidification of internal and external compartments. One family of proteins mediating Cl(-) permeability, the ClC channels, has emerged as important for all of these biological processes. The importance of ClC channels has in part been realized through studies of inherited human diseases and genetically engineered mice that display a wide range of phenotypes from kidney stones to petrified bones. These recent findings have demonstrated many eclectic functions of ClC channels and have placed Cl(-) channels in the physiological limelight.


Subject(s)
Chloride Channels/physiology , Animals , Cell Compartmentation , Cell Size , Chloride Channels/chemistry , Chloride Channels/classification , Humans , Hydrogen-Ion Concentration , Mice
11.
Curr Opin Struct Biol ; 12(4): 531-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12163078

ABSTRACT

CLC chloride channels form a large gene family that is found in bacteria, archae and eukaryotes. Previous mutagenesis studies on CLC chloride channels, combined with electrophysiology, strongly supported the theory that these channels form a homodimeric structure with one pore per subunit (a'double-barrelled' channel), and also provided clues about gating and permeation. Recently, the crystal structures of two bacterial CLC proteins have been obtained by X-ray diffraction analysis. They confirm the double-barrelled architecture, and reveal a surprisingly complex and unprecedented channel structure. At its binding site in the pore, chloride interacts with the ends of four helices that come from both sides of the membrane. A glutamate residue that protrudes into the pore is proposed to participate in gating. The structure confirms several previous conclusions from mutagenesis studies and provides an excellent framework for their interpretation.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/genetics , Ion Channel Gating/physiology , Models, Molecular , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Chloride Channels/classification , Crystallization , Crystallography, X-Ray , Cystathionine beta-Synthase/chemistry , Glutamic Acid/chemistry , Macromolecular Substances , Membrane Proteins/chemistry , Membrane Proteins/classification , Membrane Proteins/genetics , Molecular Sequence Data , Protein Binding , Protein Conformation , Structure-Activity Relationship
12.
PLoS One ; 12(9): e0184308, 2017.
Article in English | MEDLINE | ID: mdl-28886120

ABSTRACT

Chloride intracellular channel proteins (CLICs) are multi-functional proteins that are expressed in various cell types and differ in their subcellular location. Two CLIC homologs, EXL-1 (excretory canal abnormal like-1) and EXC-4 (excretory canal abnormal- 4), are encoded in the Caenorhabditis elegans genome, providing an excellent model to study the functional diversification of CLIC proteins. EXC-4 functions in excretory canal formation during normal animal development. However, to date, the physiological function of EXL-1 remains largely unknown. In this study, we demonstrate that EXL-1 responds specifically to heat stress and translocates from the cytoplasm to the nucleus in intestinal cells and body wall muscle cells under heat shock. In contrast, we do not observe EXC-4 nuclear translocation under heat shock. Full protein sequence analysis shows that EXL-1 bears a non-classic nuclear localization signal (NLS) that EXC-4 is lacking. All mammalian CLIC members have a nuclear localization signal, with the exception of CLIC3. Our phylogenetic analysis of the CLIC gene families across various animal species demonstrates that the duplication of CLICs in protostomes and deuterostomes occurred independently and that the NLS was subsequently lost in amniotes and nematodes, suggesting convergent evolution. We also observe that EXL-1 nuclear translocation occurs in a timely ordered manner in the intestine, from posterior to anterior regions. Finally, we find that exl-1 loss of function mutants are more susceptible to heat stress than wild-type animals, demonstrating functional relevance of the nuclear translocation. This research provides the first link between CLICs and environmental heat stress. We propose that C. elegans CLICs evolved to achieve different physiological functions through subcellular localization change and spatial separation in response to external or internal signals.


Subject(s)
Caenorhabditis elegans/metabolism , Chloride Channels/metabolism , Heat-Shock Response , Stress, Physiological , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Chloride Channels/chemistry , Chloride Channels/classification , Chloride Channels/genetics , Intracellular Space/metabolism , Nuclear Localization Signals , Oxidative Stress , Protein Transport
13.
Genomics Proteomics Bioinformatics ; 4(4): 253-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17531801

ABSTRACT

This study describes methods for predicting and classifying voltage-gated ion channels. Firstly, a standard support vector machine (SVM) method was developed for predicting ion channels by using amino acid composition and dipeptide composition, with an accuracy of 82.89% and 85.56%, respectively. The accuracy of this SVM method was improved from 85.56% to 89.11% when combined with PSI-BLAST similarity search. Then we developed an SVM method for classifying ion channels (potassium, sodium, calcium, and chloride) by using dipeptide composition and achieved an overall accuracy of 96.89%. We further achieved a classification accuracy of 97.78% by using a hybrid method that combines dipeptide-based SVM and hidden Markov model methods. A web server VGIchan has been developed for predicting and classifying voltage-gated ion channels using the above approaches.


Subject(s)
Calcium Channels/classification , Chloride Channels/classification , Potassium Channels, Voltage-Gated/classification , Sodium Channels/classification , Amino Acid Sequence , Dipeptides/chemistry , Ion Channel Gating , Markov Chains , Molecular Sequence Data , Sequence Analysis, Protein , Software
15.
Biochim Biophys Acta ; 1153(1): 105-10, 1993 Nov 21.
Article in English | MEDLINE | ID: mdl-8241242

ABSTRACT

We have investigated how two types of Cl- channels found in sodium transporting epithelium are regulated by arginine vasopressin (AVP). A6 cells cultured on permeable supports for 10 to 14 days have two types of Cl- channels in the apical membrane that have single channel conductances of 3 and 8 pS. In cells without AVP pretreatment, the 3 pS Cl- channel was more frequently observed than the 8 pS Cl- channel. AVP increased the open probability (Po) and single channel conductance of the 3 pS Cl- channel without significantly changing the Po or conductance of the 8 pS Cl- channels. On the other hand, AVP did not affect the number of the 3 pS Cl- channel, but increased the number of 8 pS Cl- channels. These observations suggest that AVP has two different pathways to increase apical membrane chloride conductance in distal nephron A6 cells; i.e., (1) increases the Po and single channel conductance of 3 pS Cl- channels and (2) increases the number of 8 pS Cl- channels.


Subject(s)
Arginine Vasopressin/pharmacology , Chloride Channels/drug effects , Nephrons/drug effects , Animals , Cell Line/drug effects , Cell Line/ultrastructure , Chloride Channels/classification , Electric Conductivity , Nephrons/ultrastructure , Statistics as Topic
16.
J Gen Physiol ; 122(2): 133-45, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12885875

ABSTRACT

The charge on the side chain of the internal pore residue lysine 519 (K519) of the Torpedo ClC-0 chloride (Cl-) channel affects channel conductance. Experiments that replace wild-type (WT) lysine with neutral or negatively charged residues or that modify the K519C mutant with various methane thiosulfonate (MTS) reagents show that the conductance of the channel decreases when the charge at position 519 is made more negative. This charge effect on the channel conductance diminishes in the presence of a high intracellular Cl- concentration ([Cl-]i). However, the application of high concentrations of nonpermeant ions, such as glutamate or sulfate (SO42-), does not change the conductance, suggesting that the electrostatic effects created by the charge at position 519 are unlikely due to a surface charge mechanism. Another pore residue, glutamate 127 (E127), plays an even more critical role in controlling channel conductance. This negatively charged residue, based on the structures of the homologous bacterial ClC channels, lies 4-5 A from K519. Altering the charge of this residue can influence the apparent Cl- affinity as well as the saturated pore conductance in the conductance-Cl- activity curve. Amino acid residues at the selectivity filter also control the pore conductance but mutating these residues mainly affects the maximal pore conductance. These results suggest at least two different conductance determinants in the pore of ClC-0, consistent with the most recent crystal structure of the bacterial ClC channel solved to 2.5 A, in which multiple Cl--binding sites were identified in the pore. Thus, we suggest that the occupancy of the internal Cl--binding site is directly controlled by the charged residues located at the inner pore mouth. On the other hand, the Cl--binding site at the selectivity filter controls the exit rate of Cl- and therefore determines the maximal channel conductance.


Subject(s)
Cell Membrane Permeability/physiology , Chloride Channels/chemistry , Electric Conductivity , Ion Channel Gating/physiology , Amino Acid Substitution , Animals , Chloride Channels/classification , Chloride Channels/physiology , Membrane Potentials/physiology , Point Mutation , Protein Conformation , Static Electricity , Structure-Activity Relationship , Torpedo
17.
J Gen Physiol ; 121(4): 287-300, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12668733

ABSTRACT

The distal-convoluted tubule (DCT) of the kidney absorbs NaCl mainly via an Na+-Cl- cotransporter located at the apical membrane, and Na+, K+ ATPase at the basolateral side. Cl- transport across the basolateral membrane is thought to be conductive, but the corresponding channels have not yet been characterized. In the present study, we investigated Cl- channels on microdissected mouse DCTs using the patch-clamp technique. A channel of approximately 9 pS was found in 50% of cell-attached patches showing anionic selectivity. The NPo in cell-attached patches was not modified when tubules were preincubated in the presence of 10-5 M forskolin, but the channel was inhibited by phorbol ester (10-6 M). In addition, NPo was significantly elevated when the calcium in the pipette was increased from 0 to 5 mM (NPo increased threefold), or pH increased from 6.4 to 8.0 (NPo increased 15-fold). Selectivity experiments conducted on inside-out patches showed that the Na+ to Cl- relative permeability was 0.09, and the anion selectivity sequence Cl(-)--I(-) > Br(-)--NO3(-) > F(-). Intracellular NPPB (10-4 M) and DPC (10-3 M) blocked the channel by 65% and 80%, respectively. The channel was inhibited at acid intracellular pH, but intracellular ATP and PKA had no effect. ClC-K Cl- channels are characterized by their sensitivity to the external calcium and to pH. Since immunohistochemical data indicates that ClC-K2, and perhaps ClC-K1, are present on the DCT basolateral membrane, we suggest that the channel detected in this study may belong to this subfamily of the ClC channel family.


Subject(s)
Cell Membrane/physiology , Chloride Channels/classification , Chloride Channels/physiology , Membrane Potentials/physiology , Animals , Calcium/metabolism , Cell Membrane/chemistry , Cell Membrane/drug effects , Cells, Cultured , Chloride Channels/chemistry , Colforsin/pharmacology , Electric Conductivity , Hydrogen-Ion Concentration , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Kidney Tubules, Distal , Male , Membrane Potentials/drug effects , Mice , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology
18.
Curr Pharm Des ; 11(21): 2753-64, 2005.
Article in English | MEDLINE | ID: mdl-16101453

ABSTRACT

The passage of ions to form and maintain electrochemical gradients is a key element for regulating cellular activities and is dependent on specific channel proteins or complexes. Certain ion channels have been the targets of pharmaceuticals that have had impact on a variety of cardiovascular and neurological diseases. Chloride channels regulate the movement of a major cellular anion, and in so doing they in part determine cell membrane potential, modify transepithelial transport, and maintain intracellular pH and cell volume. There are multiple families of chloride channel proteins, and respiratory, neuromuscular, and renal dysfunction may result from mutations in specific family members. Interest in chloride channels related to cancer first arose when the multidrug resistance protein (MDR/P-glycoprotein) was linked to volume-activated chloride channel activity in cancer cells from patients undergoing chemotherapy. More recently, CLC, CLIC, and CLCA intracellular chloride channels have been recognized for their contributions in modifying cell cycle, apoptosis, cell adhesion, and cell motility. Moreover, advances in structural biology and high-throughput screening provide a platform to identify chemical compounds that modulate the activities of intracellular chloride channels thereby influencing chloride ion transport and altering cell behavior. This review will focus on several chloride channel families that may contribute to the cancer phenotype and suggest how they may serve as novel targets for primary cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chloride Channels/drug effects , Chloride Channels/physiology , Amino Acid Sequence , Animals , Calcium/physiology , Cell Survival/drug effects , Cell Survival/physiology , Chloride Channels/classification , Chloride Channels/genetics , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Humans , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology
19.
Neurosci Res ; 51(1): 45-53, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15596240

ABSTRACT

The present study was performed to determine whether the voltage gated Cl- channel (CLC) expression is altered in the hippocampus of seizure sensitive (SS) gerbils, and to identify the strong fast paired-pulse inhibition in the dentate gyrus of SS gerbils is associated with altered CLC expression. In the hippocampal proper and the granule cell layer of the dentate gyrus of the SS gerbils, strong CLC-2 immunoreactivity was detected, as compared with seizure resistant (SR) gerbils. In addition, CLC-3 immunoreactivity was observed in the CA1-3 pyramidal cells, and the granule cell and the molecular layer of the dentate gyrus in the SS gerbils, whereas its immunoreactivity was rarely detected in the SR gerbils. However, CLC-3 immunoreactivity in the mossy fiber was reduced, as compared with SR gerbils. Moreover, infusion of the potential CLC inhibitor (4,4'-diisothiocyanostibene-2,2'-disulfanic acid, DIDS) reduced fast paired-pulse inhibition in the dentate gyrus of SS gerbils, although evoked responses in the dentate gyrus between SR and SS gerbils were similarly detected. These findings suggest that enhancement of CLC expression in the dentate gyrus of SS gerbils may be one of the compensatory responses for reduced GABA(A) receptor-mediated fast postsynaptic inhibitory potentials.


Subject(s)
Chloride Channels/metabolism , Dentate Gyrus/metabolism , Neural Inhibition/physiology , Seizures/physiopathology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Action Potentials , Animals , Bicuculline/pharmacology , Blotting, Western/methods , CLC-2 Chloride Channels , Chloride Channels/antagonists & inhibitors , Chloride Channels/classification , Convulsants/pharmacology , Dentate Gyrus/physiopathology , Drug Synergism , Gerbillinae , Immunohistochemistry/methods
20.
Sci Rep ; 5: 8558, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25708000

ABSTRACT

Glutamate is an indispensable neurotransmitter, triggering postsynaptic signals upon recognition by postsynaptic receptors. We questioned the phylogenetic position and the molecular details of when and where glutamate recognition arose in the glutamate-gated chloride channels. Experiments revealed that glutamate recognition requires an arginine residue in the base of the binding site, which originated at least three distinct times according to phylogenetic analysis. Most remarkably, the arginine emerged on the principal face of the binding site in the Lophotrochozoan lineage, but 65 amino acids upstream, on the complementary face, in the Ecdysozoan lineage. This combined experimental and computational approach throws new light on the evolution of synaptic signalling.


Subject(s)
Chloride Channels/metabolism , Glutamic Acid/metabolism , Amino Acid Sequence , Animals , Arginine/chemistry , Arginine/metabolism , Binding Sites , Chloride Channels/classification , Chloride Channels/genetics , Evolution, Molecular , Glutamic Acid/chemistry , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Phylogeny , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL