Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 758
Filter
1.
Nat Commun ; 15(1): 6654, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107281

ABSTRACT

The ClC-3 chloride/proton exchanger is both physiologically and pathologically critical, as it is potentiated by ATP to detect metabolic energy level and point mutations in ClC-3 lead to severe neurodegenerative diseases in human. However, why this exchanger is differentially modulated by ATP, ADP or AMP and how mutations caused gain-of-function remains largely unknow. Here we determine the high-resolution structures of dimeric wildtype ClC-3 in the apo state and in complex with ATP, ADP and AMP, and the disease-causing I607T mutant in the apo and ATP-bounded state by cryo-electron microscopy. In combination with patch-clamp recordings and molecular dynamic simulations, we reveal how the adenine nucleotides binds to ClC-3 and changes in ion occupancy between apo and ATP-bounded state. We further observe I607T mutation induced conformational changes and augments in current. Therefore, our study not only lays the structural basis of adenine nucleotides regulation in ClC-3, but also clearly indicates the target region for drug discovery against ClC-3 mediated neurodegenerative diseases.


Subject(s)
Adenosine Triphosphate , Chloride Channels , Cryoelectron Microscopy , Molecular Dynamics Simulation , Neurodegenerative Diseases , Chloride Channels/metabolism , Chloride Channels/genetics , Chloride Channels/chemistry , Humans , Adenosine Triphosphate/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Adenine Nucleotides/metabolism , Patch-Clamp Techniques , Mutation , Adenosine Diphosphate/metabolism , HEK293 Cells , Adenosine Monophosphate/metabolism , Animals , Protein Conformation
2.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39063037

ABSTRACT

The opening of the Torpedo CLC-0 chloride (Cl-) channel is known to be regulated by two gating mechanisms: fast gating and slow (common) gating. The structural basis underlying the fast-gating mechanism is better understood than that of the slow-gating mechanism, which is still largely a mystery. Our previous study on the intracellular proton (H+i)-induced inhibition of the CLC-0 anionic current led to the conclusion that the inhibition results from the slow-gate closure (also called inactivation). The conclusion was made based on substantial evidence such as a large temperature dependence of the H+i inhibition similar to that of the channel inactivation, a resistance to the H+i inhibition in the inactivation-suppressed C212S mutant, and a similar voltage dependence between the current recovery from the H+i inhibition and the recovery from the channel inactivation. In this work, we further examine the mechanism of the H+i inhibition of wild-type CLC-0 and several mutants. We observe that an anion efflux through the pore of CLC-0 accelerates the recovery from the H+i-induced inhibition, a process corresponding to the slow-gate opening. Furthermore, various inactivation-suppressed mutants exhibit different current recovery kinetics, suggesting the existence of multiple inactivated states (namely, slow-gate closed states). We speculate that protonation of the pore of CLC-0 increases the binding affinity of permeant anions in the pore, thereby generating a pore blockage of ion flow as the first step of inactivation. Subsequent complex protein conformational changes further transition the CLC-0 channel to deeper inactivated states.


Subject(s)
Chloride Channels , Ion Channel Gating , Protons , Chloride Channels/metabolism , Chloride Channels/antagonists & inhibitors , Chloride Channels/chemistry , Chloride Channels/genetics , Ion Channel Gating/drug effects , Animals , Mutation , Kinetics
3.
Methods Enzymol ; 696: 109-154, 2024.
Article in English | MEDLINE | ID: mdl-38658077

ABSTRACT

The use of molecular dynamics (MD) simulations to study biomolecular systems has proven reliable in elucidating atomic-level details of structure and function. In this chapter, MD simulations were used to uncover new insights into two phylogenetically unrelated bacterial fluoride (F-) exporters: the CLCF F-/H+ antiporter and the Fluc F- channel. The CLCF antiporter, a member of the broader CLC family, has previously revealed unique stoichiometry, anion-coordinating residues, and the absence of an internal glutamate crucial for proton import in the CLCs. Through MD simulations enhanced with umbrella sampling, we provide insights into the energetics and mechanism of the CLCF transport process, including its selectivity for F- over HF. In contrast, the Fluc F- channel presents a novel architecture as a dual topology dimer, featuring two pores for F- export and a central non-transported sodium ion. Using computational electrophysiology, we simulate the electrochemical gradient necessary for F- export in Fluc and reveal details about the coordination and hydration of both F- and the central sodium ion. The procedures described here delineate the specifics of these advanced techniques and can also be adapted to investigate other membrane protein systems.


Subject(s)
Biochemistry , Computational Biology , Fluorides , Molecular Dynamics Simulation , Fluorides/metabolism , Membrane Transport Proteins/metabolism , Ion Transport/physiology , Chloride Channels/chemistry , Chloride Channels/metabolism , Electrophysiology , Biochemistry/methods , Computational Biology/methods , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biological Transport, Active/physiology
4.
J Phys Chem B ; 128(11): 2697-2706, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38447081

ABSTRACT

CLCF fluoride/proton antiporters move fluoride ions out of bacterial cells, leading to fluoride resistance in these bacteria. However, many details about their operating mechanisms remain unclear. Here, we report a combined quantum-mechanical/molecular-mechanical (QM/MM) study of a CLCF homologue from Enterococci casseliflavus (Eca), in accord with the previously proposed windmill mechanism. Our multiscale modeling sheds light on two critical steps in the transport cycle: (i) the external gating residue E118 pushing a fluoride in the external binding site into the extracellular vestibule and (ii) an incoming fluoride reconquering the external binding site by forcing out E118. Both steps feature competitions for the external binding site between the negatively charged carboxylate of E118 and the fluoride. Remarkably, the displaced E118 by fluoride accepts a proton from the nearby R117, initiating the next transport cycle. We also demonstrate the importance of accurate quantum descriptions of fluoride solvation. Our results provide clues to the mysterious E318 residue near the central binding site, suggesting that the transport activities are unlikely to be disrupted by the glutamate interacting with a well-solvated fluoride at the central binding site. This differs significantly from the structurally similar CLC chloride/proton antiporters, where a fluoride trapped deep in the hydrophobic pore causes the transporter to be locked down. A free-energy barrier of 10-15 kcal/mol was estimated via umbrella sampling for a fluoride ion traveling through the pore to repopulate the external binding site.


Subject(s)
Antiporters , Protons , Antiporters/chemistry , Antiporters/metabolism , Fluorides/chemistry , Models, Molecular , Membrane Transport Proteins/metabolism , Chlorides/chemistry , Chloride Channels/chemistry , Chloride Channels/metabolism , Ion Transport
5.
J Am Chem Soc ; 146(7): 4665-4679, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38319142

ABSTRACT

The dysfunction and defects of ion channels are associated with many human diseases, especially for loss-of-function mutations in ion channels such as cystic fibrosis transmembrane conductance regulator mutations in cystic fibrosis. Understanding ion channels is of great current importance for both medical and fundamental purposes. Such an understanding should include the ability to predict mutational effects and describe functional and mechanistic effects. In this work, we introduce an approach to predict mutational effects based on kinetic information (including reaction barriers and transition state locations) obtained by studying the working mechanism of target proteins. Specifically, we take the Ca2+-activated chloride channel TMEM16A as an example and utilize the computational biology model to predict the mutational effects of key residues. Encouragingly, we verified our predictions through electrophysiological experiments, demonstrating a 94% prediction accuracy regarding mutational directions. The mutational strength assessed by Pearson's correlation coefficient is -0.80 between our calculations and the experimental results. These findings suggest that the proposed methodology is reliable and can provide valuable guidance for revealing functional mechanisms and identifying key residues of the TMEM16A channel. The proposed approach can be extended to a broad scope of biophysical systems.


Subject(s)
Chloride Channels , Chlorides , Humans , Chlorides/metabolism , Anoctamin-1/genetics , Anoctamin-1/metabolism , Chloride Channels/genetics , Chloride Channels/chemistry , Chloride Channels/metabolism , Mutation , Signal Transduction , Calcium/metabolism
6.
Sci Adv ; 9(41): eadg4479, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37831762

ABSTRACT

ClC-6 is a late endosomal voltage-gated chloride-proton exchanger that is predominantly expressed in the nervous system. Mutated forms of ClC-6 are associated with severe neurological disease. However, the mechanistic role of ClC-6 in normal and pathological states remains largely unknown. Here, we present cryo-EM structures of ClC-6 that guided subsequent functional studies. Previously unrecognized ATP binding to cytosolic ClC-6 domains enhanced ion transport activity. Guided by a disease-causing mutation (p.Y553C), we identified an interaction network formed by Y553/F317/T520 as potential hotspot for disease-causing mutations. This was validated by the identification of a patient with a de novo pathogenic variant p.T520A. Extending these findings, we found contacts between intramembrane helices and connecting loops that modulate the voltage dependence of ClC-6 gating and constitute additional candidate regions for disease-associated gain-of-function mutations. Besides providing insights into the structure, function, and regulation of ClC-6, our work correctly predicts hotspots for CLCN6 mutations in neurodegenerative disorders.


Subject(s)
Chloride Channels , Neurodegenerative Diseases , Humans , Chloride Channels/chemistry , Chloride Channels/genetics , Ion Transport , Mutation , Neurodegenerative Diseases/genetics , Structure-Activity Relationship
7.
J Microbiol Biotechnol ; 33(7): 857-863, 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37100762

ABSTRACT

Pathogenic bacteria that colonize the human intestinal tract have evolved strategies to overcome acidic conditions when they pass through the gastrointestinal tract. Amino acid-mediated acid resistance systems are effective survival strategies in a stomach that is full of amino acid substrate. The amino acid antiporter, amino acid decarboxylase, and ClC chloride antiporter are all engaged in these systems, and each one plays a role in protecting against or adapting to the acidic environment. The ClC chloride antiporter, a member of the ClC channel family, eliminates negatively charged intracellular chloride ions to avoid inner membrane hyperpolarization as an electrical shunt of the acid resistance system. In this review, we will discuss the structure and function of the prokaryotic ClC chloride antiporter of amino acid-mediated acid resistance system.


Subject(s)
Antiporters , Chlorides , Humans , Chlorides/metabolism , Antiporters/chemistry , Antiporters/metabolism , Amino Acids , Gram-Negative Bacteria/metabolism , Chloride Channels/chemistry , Chloride Channels/metabolism
8.
Adv Exp Med Biol ; 1422: 279-304, 2023.
Article in English | MEDLINE | ID: mdl-36988885

ABSTRACT

Chloride fluxes through homo-dimeric calcium-activated channels TMEM16A and TMEM16B are critical to blood pressure, gastrointestinal motility, hormone, fluid and electrolyte secretion, pain sensation, sensory transduction, and neuronal and muscle excitability. Their gating depends on the voltage-dependent binding of two intracellular calcium ions to a high-affinity site formed by acidic residues from α-helices 6-8 in each monomer. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), a low-abundant lipid of the inner leaflet, supports TMEM16A function; it allows TMEM16A to evade the down-regulation induced by calcium, poly-L-lysine, or PI(4,5)P2 5-phosphatase. In stark contrast, adding or removing PI(4,5)P2 diminishes or increases TMEM16B function, respectively. PI(4,5)P2-binding sites on TMEM16A, and presumably on TMEM16B, are on the cytosolic side of α-helices 3-5, opposite the calcium-binding sites. This modular structure suggested that PI(4,5)P2 and calcium cooperate to maintain the conductive state in TMEM16A. Cholesterol, the second-largest constituent of the plasma membrane, also regulates TMEM16A though the mechanism, functional outcomes, binding site(s), and effects on TMEM16A and TMEM16B remain unknown.


Subject(s)
Chloride Channels , Phosphatidylinositols , Humans , Chloride Channels/genetics , Chloride Channels/chemistry , Chloride Channels/metabolism , Anoctamin-1/metabolism , Calcium/metabolism , Cholesterol , Calcium Channels , HEK293 Cells
9.
Cardiovasc Pathol ; 65: 107525, 2023.
Article in English | MEDLINE | ID: mdl-36781068

ABSTRACT

Transmembrane protein 16A (TMEM16A), a member of the TMEM16 family, is the molecular basis of Ca2+-activated chloride channels (CaCCs) and is involved in a variety of physiological and pathological processes. Previous studies have focused more on respiratory-related diseases and tumors. However, recent studies have identified an important role for TMEM16A in cardiovascular diseases, especially in pulmonary hypertension. TMEM16A is expressed in both pulmonary artery smooth muscle cells and pulmonary artery endothelial cells and is involved in the development of pulmonary hypertension. This paper presents the structure and function of TMEM16A, the pathogenesis of pulmonary hypertension, and highlights the role and mechanism of TMEM16A in pulmonary hypertension, summarizing the controversies in this field and taking into account hypertension and portal hypertension, which have similar pathogenesis. It is hoped that the unique role of TMEM16A in pulmonary hypertension will be illustrated and provide ideas for research in this area.


Subject(s)
Hypertension, Pulmonary , Hypertension , Humans , Anoctamin-1 , Endothelial Cells/metabolism , Chloride Channels/genetics , Chloride Channels/chemistry , Chloride Channels/metabolism , Hypertension/pathology
10.
Mol Phylogenet Evol ; 177: 107595, 2022 12.
Article in English | MEDLINE | ID: mdl-35914647

ABSTRACT

Most of Transmembrane protein 16 (TMEM16) family members function as either a Ca2+-activated Cl- channel (CaCC) or phospholipid scramblase (CaPLSase) and play diverse physiological roles. It is well conserved in eukaryotes; however, the origin and evolution of different subfamilies in Metazoa are not yet understood. To uncover the evolutionary history of the TMEM16 family, we analyzed 398 proteins from 74 invertebrate species using evolutionary genomics. We found that the TMEM16C-F and J subfamilies are vertebrate-specific, but the TMEM16A/B, G, H, and K subfamilies are ancient and present in many, but not all metazoan species. The most ancient subfamilies in Metazoa, TMEM16L and M, are only maintained in limited species. TMEM16N and O are Cnidaria- and Ecdysozoa-specific subfamilies, respectively, and Ctenophora, Xenacoelomorpha, and Rotifera contain species-specific proteins. We also identified TMEM16 genes that are closely linked together in the genome, suggesting that they have been generated via recent gene duplication. The anoctamin domain structures of invertebrate-specific TMEM16 proteins predicted by AlphaFold2 contain conserved Ca2+-binding motifs and permeation pathways with either narrow or wide inner gates. The inner gate distance of TMEM16 protein may have frequently switched during metazoan evolution, and thus determined the function of the protein as either CaCC or CaPLSase. These results demonstrate that TMEM16 family has evolved by gene gain and loss in metazoans, and the genes have been generally under purifying selection to maintain protein structures and physiological functions.


Subject(s)
Anoctamins , Phospholipid Transfer Proteins , Animals , Anoctamins/genetics , Anoctamins/metabolism , Chloride Channels/chemistry , Chloride Channels/genetics , Chloride Channels/metabolism , Eukaryota/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Phylogeny
11.
J Gen Physiol ; 154(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35687042

ABSTRACT

Numerous essential physiological processes depend on the TMEM16A-mediated Ca2+-activated chloride fluxes. Extensive structure-function studies have helped to elucidate the Ca2+ gating mechanism of TMEM16A, revealing a Ca2+-sensing element close to the anion pore that alters conduction. However, substrate selection and the substrate-gating relationship in TMEM16A remain less explored. Here, we study the gating-permeant anion relationship on mouse TMEM16A expressed in HEK 293 cells using electrophysiological recordings coupled with site-directed mutagenesis. We show that the apparent Ca2+ sensitivity of TMEM16A increased with highly permeant anions and SCN- mole fractions, likely by stabilizing bound Ca2+. Conversely, mutations at crucial gating elements, including the Ca2+-binding site 1, the transmembrane helix 6 (TM6), and the hydrophobic gate, impaired the anion permeability and selectivity of TMEM16A. Finally, we found that, unlike anion-selective wild-type channels, the voltage dependence of unselective TMEM16A mutant channels was less sensitive to SCN-. Therefore, our work identifies structural determinants of selectivity at the Ca2+ site, TM6, and hydrophobic gate and reveals a reciprocal regulation of gating and selectivity. We suggest that this regulation is essential to set ionic selectivity and the Ca2+ and voltage sensitivities in TMEM16A.


Subject(s)
Calcium , Chloride Channels , Animals , Anions/metabolism , Anoctamin-1/genetics , Calcium/metabolism , Chloride Channels/chemistry , Chloride Channels/genetics , HEK293 Cells , Humans , Ion Channel Gating , Mice , Neoplasm Proteins/metabolism
12.
Article in English | MEDLINE | ID: mdl-35202806

ABSTRACT

The calcium-activated chloride channel TMEM16A (ANO1) supports the passive movement of chloride ions across membranes and controls critical cell functions. Here we study the block of wild-type and mutant TMEM16A channels expressed in HEK293 cells by oleic acid, a monounsaturated omega-9 fatty acid beneficial for cardiovascular health. We found that oleic acid irreversibly blocks TMEM16A in a dose- and voltage-dependent manner at low intracellular Ca2+. We tested whether oleic acid interacted with the TMEM16A pore, varying the permeant anion concentration and mutating pore residues. Lowering the permeating anion concentration in the intracellular side did nothing but the blockade was intensified by increasing the anion concentration in the extracellular side. However, the blockade of the pore mutants E633A and I641A was voltage-independent, and the I641A IC50, a mutant with the inner hydrophobic gate in disarray, increased 16-fold. Furthermore, the uncharged methyl-oleate blocked 20-24% of the wild-type and I641A channels regardless of voltage. Our findings suggest that oleic acid inhibits TMEM16A by an allosteric mechanism after the electric field drives oleic acid's charged moiety inside the pore. Block of TMEM16A might be why oleic acid has a beneficial impact on the cardiovascular system.


Subject(s)
Chloride Channels , Oleic Acid , Anions/metabolism , Anoctamin-1/genetics , Anoctamin-1/metabolism , Calcium/metabolism , Chloride Channels/chemistry , Chloride Channels/genetics , Chloride Channels/metabolism , HEK293 Cells , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oleic Acid/pharmacology
13.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35197289

ABSTRACT

Light-driven chloride-pumping rhodopsins actively transport anions, including various halide ions, across cell membranes. Recent studies using time-resolved serial femtosecond crystallography (TR-SFX) have uncovered the structural changes and ion transfer mechanisms in light-driven cation-pumping rhodopsins. However, the mechanism by which the conformational changes pump an anion to achieve unidirectional ion transport, from the extracellular side to the cytoplasmic side, in anion-pumping rhodopsins remains enigmatic. We have collected TR-SFX data of Nonlabens marinus rhodopsin-3 (NM-R3), derived from a marine flavobacterium, at 10-µs and 1-ms time points after photoexcitation. Our structural analysis reveals the conformational alterations during ion transfer and after ion release. Movements of the retinal chromophore initially displace a conserved tryptophan to the cytoplasmic side of NM-R3, accompanied by a slight shift of the halide ion bound to the retinal. After ion release, the inward movements of helix C and helix G and the lateral displacements of the retinal block access to the extracellular side of NM-R3. Anomalous signal data have also been obtained from NM-R3 crystals containing iodide ions. The anomalous density maps provide insight into the halide binding site for ion transfer in NM-R3.


Subject(s)
Chloride Channels/chemistry , Lasers , Chloride Channels/metabolism , Crystallography , Cytoplasm/metabolism , Ion Transport , Light , Protein Conformation , X-Rays
14.
Nat Commun ; 12(1): 6913, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824283

ABSTRACT

Tweety homologs (TTYHs) comprise a conserved family of transmembrane proteins found in eukaryotes with three members (TTYH1-3) in vertebrates. They are widely expressed in mammals including at high levels in the nervous system and have been implicated in cancers and other diseases including epilepsy, chronic pain, and viral infections. TTYHs have been reported to form Ca2+- and cell volume-regulated anion channels structurally distinct from any characterized protein family with potential roles in cell adhesion, migration, and developmental signaling. To provide insight into TTYH family structure and function, we determined cryo-EM structures of Mus musculus TTYH2 and TTYH3 in lipid nanodiscs. TTYH2 and TTYH3 adopt a previously unobserved fold which includes an extended extracellular domain with a partially solvent exposed pocket that may be an interaction site for hydrophobic molecules. In the presence of Ca2+, TTYH2 and TTYH3 form homomeric cis-dimers bridged by extracellularly coordinated Ca2+. Strikingly, in the absence of Ca2+, TTYH2 forms trans-dimers that span opposing membranes across a ~130 Å intermembrane space as well as a monomeric state. All TTYH structures lack ion conducting pathways and we do not observe TTYH2-dependent channel activity in cells. We conclude TTYHs are not pore forming subunits of anion channels and their function may involve Ca2+-dependent changes in quaternary structure, interactions with hydrophobic molecules near the extracellular membrane surface, and/or association with additional protein partners.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/metabolism , Dimerization , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Animals , Anoctamins/chemistry , Biological Transport , Calcium/metabolism , Cell Adhesion , Cell Size , Chloride Channels/genetics , Chronic Pain , Cryoelectron Microscopy , Eukaryota , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/genetics , Mice , Receptor, EphB2 , Signal Transduction
15.
Nat Commun ; 12(1): 4893, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385445

ABSTRACT

The Tweety homologs (TTYHs) are members of a conserved family of eukaryotic membrane proteins that are abundant in the brain. The three human paralogs were assigned to function as anion channels that are either activated by Ca2+ or cell swelling. To uncover their unknown architecture and its relationship to function, we have determined the structures of human TTYH1-3 by cryo-electron microscopy. All structures display equivalent features of a dimeric membrane protein that contains five transmembrane segments and an extended extracellular domain. As none of the proteins shows attributes reminiscent of an anion channel, we revisited functional experiments and did not find any indication of ion conduction. Instead, we find density in an extended hydrophobic pocket contained in the extracellular domain that emerges from the lipid bilayer, which suggests a role of TTYH proteins in the interaction with lipid-like compounds residing in the membrane.


Subject(s)
Chloride Channels/ultrastructure , Cryoelectron Microscopy/methods , Membrane Proteins/ultrastructure , Neoplasm Proteins/ultrastructure , Chloride Channels/chemistry , Chloride Channels/metabolism , Humans , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/ultrastructure , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Protein Binding , Protein Conformation
16.
Sci Rep ; 11(1): 13127, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162897

ABSTRACT

The widely expressed two-pore homodimeric inward rectifier CLC-2 chloride channel regulates transepithelial chloride transport, extracellular chloride homeostasis, and neuronal excitability. Each pore is independently gated at hyperpolarized voltages by a conserved pore glutamate. Presumably, exiting chloride ions push glutamate outwardly while external protonation stabilizes it. To understand the mechanism of mouse CLC-2 opening we used homology modelling-guided structure-function analysis. Structural modelling suggests that glutamate E213 interacts with tyrosine Y561 to close a pore. Accordingly, Y561A and E213D mutants are activated at less hyperpolarized voltages, re-opened at depolarized voltages, and fast and common gating components are reduced. The double mutant cycle analysis showed that E213 and Y561 are energetically coupled to alter CLC-2 gating. In agreement, the anomalous mole fraction behaviour of the voltage dependence, measured by the voltage to induce half-open probability, was strongly altered in these mutants. Finally, cytosolic acidification or high extracellular chloride concentration, conditions that have little or no effect on WT CLC-2, induced reopening of Y561 mutants at positive voltages presumably by the inward opening of E213. We concluded that the CLC-2 gate is formed by Y561-E213 and that outward permeant anions open the gate by electrostatic and steric interactions.


Subject(s)
Chloride Channels/chemistry , Ion Channel Gating , Amino Acid Sequence , Animals , CLC-2 Chloride Channels , Cattle , Chloride Channels/genetics , Chloride Channels/metabolism , Chlorides/metabolism , Humans , Mice , Mutation , Protein Structure, Tertiary , Sequence Alignment , Structure-Activity Relationship
17.
Nature ; 594(7863): 385-390, 2021 06.
Article in English | MEDLINE | ID: mdl-34135520

ABSTRACT

Understanding structural dynamics of biomolecules at the single-molecule level is vital to advancing our knowledge of molecular mechanisms. Currently, there are few techniques that can capture dynamics at the sub-nanometre scale and in physiologically relevant conditions. Atomic force microscopy (AFM)1 has the advantage of analysing unlabelled single molecules in physiological buffer and at ambient temperature and pressure, but its resolution limits the assessment of conformational details of biomolecules2. Here we present localization AFM (LAFM), a technique developed to overcome current resolution limitations. By applying localization image reconstruction algorithms3 to peak positions in high-speed AFM and conventional AFM data, we increase the resolution beyond the limits set by the tip radius, and resolve single amino acid residues on soft protein surfaces in native and dynamic conditions. LAFM enables the calculation of high-resolution maps from either images of many molecules or many images of a single molecule acquired over time, facilitating single-molecule structural analysis. LAFM is a post-acquisition image reconstruction method that can be applied to any biomolecular AFM dataset.


Subject(s)
Microscopy, Atomic Force/methods , Microscopy, Atomic Force/standards , Algorithms , Amino Acids/chemistry , Annexin A5/chemistry , Annexin A5/ultrastructure , Aquaporins/chemistry , Aquaporins/ultrastructure , Chloride Channels/chemistry , Chloride Channels/ultrastructure , Datasets as Topic , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/ultrastructure , Humans , Hydrogen-Ion Concentration , Molecular Dynamics Simulation
18.
Int J Mol Sci ; 22(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33806823

ABSTRACT

Sperm motility is linked to the activation of signaling pathways that trigger movement. These pathways are mainly dependent on Ca2+, which acts as a secondary messenger. The maintenance of adequate Ca2+ concentrations is possible thanks to proper concentrations of other ions, such as K+ and Na+, among others, that modulate plasma membrane potential and the intracellular pH. Like in every cell, ion homeostasis in spermatozoa is ensured by a vast spectrum of ion channels supported by the work of ion pumps and transporters. To achieve success in fertilization, sperm ion channels have to be sensitive to various external and internal factors. This sensitivity is provided by specific channel structures. In addition, novel sperm-specific channels or isoforms have been found with compositions that increase the chance of fertilization. Notably, the most significant sperm ion channel is the cation channel of sperm (CatSper), which is a sperm-specific Ca2+ channel required for the hyperactivation of sperm motility. The role of other ion channels in the spermatozoa, such as voltage-gated Ca2+ channels (VGCCs), Ca2+-activated Cl-channels (CaCCs), SLO K+ channels or voltage-gated H+ channels (VGHCs), is to ensure the activation and modulation of CatSper. As the activation of sperm motility differs among metazoa, different ion channels may participate; however, knowledge regarding these channels is still scarce. In the present review, the roles and structures of the most important known ion channels are described in regard to regulation of sperm motility in animals.


Subject(s)
Ion Channel Gating , Ion Channels/chemistry , Ion Channels/metabolism , Sperm Motility , Spermatozoa/physiology , Animals , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Chloride Channels/chemistry , Chloride Channels/genetics , Chloride Channels/metabolism , Humans , Ion Channels/genetics , Male , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels/metabolism , Sodium Channels/chemistry , Sodium Channels/genetics , Sodium Channels/metabolism , Structure-Activity Relationship
19.
Biomed Pharmacother ; 138: 111407, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33765585

ABSTRACT

Epithelial ovarian cancers (EOC) present as malignant tumors with high mortality in the female reproductive system diseases. Acquired resistance to paclitaxel (PTX), one of the first-line treatment of EOC, remains a therapeutic challenge. ClC-3, a member of the voltage-gated Cl- channels, plays an essential role in a variety of cellular activities, including chemotherapeutic resistance. Here, we demonstrated that the protein expression and channel function of ClC-3 was upregulated in PTX resistance A2780/PTX cells compared with its parental A2780 cells. The silence of ClC-3 expression by siRNA in A2780/PTX cells partly recovered the PTX sensitivity through restored the G2/M arrest and resumed the chloride channel blocked. ClC-3 siRNA both inhibited the expression of ClC-3 and ß-tubulin, whereas the ß-tubulin siRNA reduced the expression of itself only, without affecting the expression of ClC-3. Moreover, treatment of ClC-3 siRNA in A2780/PTX cells increased the polymerization ratio of ß-tubulin, and the possibility of proteins interaction between ClC-3 and ß-tubulin was existing. Take together, the over-expression of ClC-3 protein in PTX-resistance ovarian cancer cells promotes the combination of ClC-3 and ß-tubulin, which in turn increase the ration of free form and decrease the quota of the polymeric form of ß-tubulin, and finally reduce the sensitivity to PTX. Our findings elucidated a novel function of ClC-3 in regulating PTX resistance and ClC-3 could serve as a potential target to overcome the PTX resistance ovarian cancer.


Subject(s)
Chloride Channels/biosynthesis , Drug Resistance, Neoplasm/drug effects , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Tubulin Modulators/metabolism , Tubulin/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Chloride Channels/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/physiology , Female , Humans , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Polymerization/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary
20.
Mol Neurobiol ; 58(6): 2990-2999, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33590434

ABSTRACT

Vesicular chloride/proton exchangers of the CLC family are critically involved in the function of the endosomal-lysosomal pathway. Their dysfunction leads to severe disorders including intellectual disability and epilepsy for ClC-4, Dent's disease for ClC-5, and lysosomal storage disease and osteopetrosis for ClC-7. Here, we report a de novo variant p.Glu200Ala (p.E200A; c.599A>C) of the late endosomal ClC-6, encoded by CLCN6, in a patient with West syndrome (WS), severe developmental delay, autism, movement disorder, microcephaly, facial dysmorphism, and visual impairment. Mutation of this conserved glutamate uncouples chloride transport from proton antiport by ClC-6. This affects organellar ion homeostasis and was shown to be deleterious for other CLCs. In this study, we found that upon heterologous expression, the ClC-6 E200A variant caused autophagosome accumulation and impaired the clearance of autophagosomes by blocking autophagosome-lysosome fusion. Our study provides clinical and functional support for an association between CLCN6 variants and WS. Our findings also provide novel insights into the molecular mechanisms underlying the pathogenesis of WS, suggesting an involvement of autophagic-lysosomal dysfunction.


Subject(s)
Autophagy/genetics , Chloride Channels/genetics , Chlorides/metabolism , Lysosomes/metabolism , Mutation/genetics , Protons , Spasms, Infantile/genetics , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Chloride Channels/chemistry , Computer Simulation , HEK293 Cells , HeLa Cells , Humans , Infant , Infant, Newborn , Male , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL