Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
Science ; 349(6248): 647-50, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26113638

ABSTRACT

Light-gated rhodopsin cation channels from chlorophyte algae have transformed neuroscience research through their use as membrane-depolarizing optogenetic tools for targeted photoactivation of neuron firing. Photosuppression of neuronal action potentials has been limited by the lack of equally efficient tools for membrane hyperpolarization. We describe anion channel rhodopsins (ACRs), a family of light-gated anion channels from cryptophyte algae that provide highly sensitive and efficient membrane hyperpolarization and neuronal silencing through light-gated chloride conduction. ACRs strictly conducted anions, completely excluding protons and larger cations, and hyperpolarized the membrane of cultured animal cells with much faster kinetics at less than one-thousandth of the light intensity required by the most efficient currently available optogenetic proteins. Natural ACRs provide optogenetic inhibition tools with unprecedented light sensitivity and temporal precision.


Subject(s)
Chloride Channels/physiology , Cryptophyta/metabolism , Membrane Potentials/radiation effects , Neurons/radiation effects , Optogenetics/methods , Rhodopsins, Microbial/physiology , Amino Acid Sequence , Chloride Channels/classification , Chloride Channels/genetics , Cryptophyta/genetics , HEK293 Cells , Humans , Ion Channel Gating , Light , Membrane Potentials/physiology , Molecular Sequence Data , Neural Inhibition , Neurons/physiology , Photic Stimulation , Phylogeny , Rhodopsins, Microbial/classification , Rhodopsins, Microbial/genetics , Transfection
5.
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
6.
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
8.
Compr Physiol ; 1(4): 2155-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-23733701

ABSTRACT

Ca(2+)-activated Cl(-) channels (CaCCs) are plasma membrane proteins involved in various important physiological processes. In epithelial cells, CaCC activity mediates the secretion of Cl(-) and of other anions, such as bicarbonate and thiocyanate. In smooth muscle and excitable cells of the nervous system, CaCCs have an excitatory role coupling intracellular Ca(2+) elevation to membrane depolarization. Recent studies indicate that TMEM16A (transmembrane protein 16 A or anoctamin 1) and TMEM16B (transmembrane protein 16 B or anoctamin 2) are CaCC-forming proteins. Induced expression of TMEM16A and B in null cells by transfection causes the appearance of Ca(2+)-activated Cl(-) currents similar to those described in native tissues. Furthermore, silencing of TMEM16A by RNAi causes disappearance of CaCC activity in cells from airway epithelium, biliary ducts, salivary glands, and blood vessel smooth muscle. Mice devoid of TMEM16A expression have impaired Ca(2+)-dependent Cl(-) secretion in the epithelial cells of the airways, intestine, and salivary glands. These animals also show a loss of gastrointestinal motility, a finding consistent with an important function of TMEM16A in the electrical activity of gut pacemaker cells, that is, the interstitial cells of Cajal. Identification of TMEM16 proteins will help to elucidate the molecular basis of Cl(-) transport.


Subject(s)
Chloride Channels/metabolism , Chlorides/metabolism , Animals , Chloride Channels/classification , Chloride Channels/genetics , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Gene Expression , Humans , Ion Transport , Muscle Cells/metabolism , Neurons/metabolism , Organ Specificity
9.
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
10.
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
11.
FEBS Lett ; 584(10): 2093-101, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20085760

ABSTRACT

Chloride intracellular channel proteins (CLICs) are distinct from most ion channels in that they have both soluble and integral membrane forms. CLICs are highly conserved in chordates, with six vertebrate paralogues. CLIC-like proteins are found in other metazoans. CLICs form channels in artificial bilayers in a process favoured by oxidising conditions and low pH. They are structurally plastic, with CLIC1 adopting two distinct soluble conformations. Phylogenetic and structural data indicate that CLICs are likely to have enzymatic function. The physiological role of CLICs appears to be maintenance of intracellular membranes, which is associated with tubulogenesis but may involve other substructures.


Subject(s)
Chloride Channels/metabolism , Enzymes/metabolism , Animals , Cell Membrane/metabolism , Chloride Channels/chemistry , Chloride Channels/classification , Chloride Channels/genetics , Cytoskeleton/metabolism , Enzymes/chemistry , Enzymes/classification , Enzymes/genetics , Humans , Hydrogen-Ion Concentration , Oxidation-Reduction
12.
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
13.
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
14.
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
15.
Nat Rev Drug Discov ; 8(2): 153-71, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19153558

ABSTRACT

Chloride channels represent a relatively under-explored target class for drug discovery as elucidation of their identity and physiological roles has lagged behind that of many other drug targets. Chloride channels are involved in a wide range of biological functions, including epithelial fluid secretion, cell-volume regulation, neuroexcitation, smooth-muscle contraction and acidification of intracellular organelles. Mutations in several chloride channels cause human diseases, including cystic fibrosis, macular degeneration, myotonia, kidney stones, renal salt wasting and hyperekplexia. Chloride-channel modulators have potential applications in the treatment of some of these disorders, as well as in secretory diarrhoeas, polycystic kidney disease, osteoporosis and hypertension. Modulators of GABA(A) (gamma-aminobutyric acid A) receptor chloride channels are in clinical use and several small-molecule chloride-channel modulators are in preclinical development and clinical trials. Here, we discuss the broad opportunities that remain in chloride-channel-based drug discovery.


Subject(s)
Chloride Channels/physiology , Chlorides , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Drug Delivery Systems/methods , Ion Channel Gating/physiology , Chloride Channels/antagonists & inhibitors , Chloride Channels/classification , Chloride Channels/drug effects , Chlorides/metabolism , Chlorides/physiology , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Humans , Ion Channel Gating/drug effects
16.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 25(4): 980-3, 2008 Aug.
Article in Chinese | 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
17.
Dev Dyn ; 237(9): 2566-74, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18729231

ABSTRACT

The TMEM16 protein family has recently been identified through several different experimental strategies including bioinformatic and microarray-based approaches. In mice and humans, there exist 10 paralogs with each containing eight putative transmembrane domains and a conserved C-terminal domain of unknown function. Mutation of at least one member of this family is associated with a human disorder, and several members of this gene family are overexpressed in different types of cancer. Despite their apparent relevance to normal development and disease, little is known about the expression of TMEM16 paralogs during embryonic development. Here, we provide a phylogenetic analysis of mouse and human TMEM16 paralogs and report the expression of Tmem16a, Tmem16b, Tmem16c, Tmem16f, Tmem16h, Tmem16j, and Tmem16k during murine embryogenesis with an emphasis on the respiratory, digestive, skeletal, and integumentary systems. These data should encourage investigations into the functions of TMEM16 paralogs in vertebrate development.


Subject(s)
Chloride Channels/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Amino Acid Sequence , Animals , Anoctamin-1 , Chloride Channels/classification , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , In Situ Hybridization , Mice , Molecular Sequence Data , Phylogeny , Respiratory System/embryology , Respiratory System/metabolism , Sequence Homology, Amino Acid , Skin/embryology , Skin/metabolism
18.
Eur J Neurosci ; 26(8): 2119-30, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17927776

ABSTRACT

The shape and volume of microglia (brain immune cells) change when they activate during brain inflammation and become migratory and phagocytic. Swollen rat microglia express a large Cl(-) current (I(Clswell)), whose biophysical properties and functional roles are poorly understood and whose molecular identity is unknown. We constructed a fingerprint of useful biophysical properties for comparison with I(Clswell) in other cell types and with cloned Cl(-) channels. The microglial I(Clswell) was rapidly activated by cell swelling but not by voltage, and showed no time-dependence during voltage-clamp steps. Like I(Clswell) in many cell types, the halide selectivity sequence was I(-) > Br(-) > Cl(-) > F(-). However, it differed in lacking inactivation, even at +100 mV with high extracellular Mg(2+), and in having a much lower single-channel conductance: 1-3 pS. Based on these fundamental differences, the microglia channel is apparently a different gene product than the more common intermediate-conductance I(Clswell). Microglia express several candidate genes, with relative mRNA expression levels of: CLIC1 > ClC3 > I(Cln) > or = ClC2 > Best2 > Best1 > or = Best3 > Best4. Using a pharmacological toolbox, we show that all drugs that reduced the microglia current (NPPB, IAA-94, flufenamic acid and DIOA) increased the resting cell volume in isotonic solution and inhibited the regulatory volume decrease that followed cell swelling in hypotonic solution. Both channel blockers tested (NPPB and flufenamic acid) dose-dependently inhibited microglia phagocytosis of E. coli bacteria. Because I(Clswell) is involved in microglia functions that involve shape and volume changes, it is potentially important for controlling their ability to migrate to damage sites and phagocytose dead cells and debris.


Subject(s)
Chloride Channels/metabolism , Microglia/physiology , Phagocytosis/physiology , Analysis of Variance , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Chelating Agents/pharmacology , Chloride Channels/classification , Chloride Channels/genetics , Dose-Response Relationship, Drug , Flow Cytometry/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hypotonic Solutions/pharmacology , Magnesium/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microglia/drug effects , Patch-Clamp Techniques/methods , Phagocytosis/drug effects , Rats , Rats, Wistar
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...