Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO J ; 42(24): e115030, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-37984335

RESUMEN

Agonist binding in ligand-gated ion channels is coupled to structural rearrangements around the binding site, followed by the opening of the channel pore. In this process, agonist efficacy describes the equilibrium between open and closed conformations in a fully ligand-bound state. Calcium-activated chloride channels in the TMEM16 family are important sensors of intracellular calcium signals and are targets for pharmacological modulators, yet a mechanistic understanding of agonist efficacy has remained elusive. Using a combination of cryo-electron microscopy, electrophysiology, and autocorrelation analysis, we now show that agonist efficacy in the ligand-gated channel TMEM16A is dictated by the conformation of the pore-lining helix α6 around the Ca2+ -binding site. The closure of the binding site, which involves the formation of a π-helix below a hinge region in α6, appears to be coupled to the opening of the inner pore gate, thereby governing the channel's open probability and conductance. Our results provide a mechanism for agonist binding and efficacy and a structural basis for the design of potentiators and partial agonists in the TMEM16 family.


Asunto(s)
Canales de Cloruro , Activación del Canal Iónico , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Anoctamina-1/genética , Anoctamina-1/química , Anoctamina-1/metabolismo , Ligandos , Microscopía por Crioelectrón , Sitios de Unión , Calcio/metabolismo
2.
J Biol Chem ; 299(6): 104780, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37142220

RESUMEN

The calcium-activated chloride channel TMEM16A is a potential drug target to treat hypertension, secretory diarrhea, and several cancers. However, all reported TMEM16A structures are either closed or desensitized, and direct inhibition of the open state by drug molecules lacks a reliable structural basis. Therefore, revealing the druggable pocket of TMEM16A exposed in the open state is important for understanding protein-ligand interactions and facilitating rational drug design. Here, we reconstructed the calcium-activated open conformation of TMEM16A using an enhanced sampling algorithm and segmental modeling. Furthermore, we identified an open-state druggable pocket and screened a potent TMEM16A inhibitor, etoposide, which is a derivative of a traditional herbal monomer. Molecular simulations and site-directed mutagenesis showed that etoposide binds to the open state of TMEM16A, thereby blocking the ion conductance pore of the channel. Finally, we demonstrated that etoposide can target TMEM16A to inhibit the proliferation of prostate cancer PC-3 cells. Together, these findings provide a deep understanding of the TMEM16A open state at an atomic level and identify pockets for the design of novel inhibitors with broad applications in chloride channel biology, biophysics, and medicinal chemistry.


Asunto(s)
Anoctamina-1 , Modelos Moleculares , Humanos , Masculino , Anoctamina-1/química , Anoctamina-1/metabolismo , Calcio/metabolismo , Etopósido/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Simulación por Computador
3.
Biochim Biophys Acta Biomembr ; 1864(1): 183777, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34537214

RESUMEN

TMEM16A, the calcium-activated chloride channel, is broadly expressed and plays pivotal roles in diverse physiological processes. To understand the structural and functional relationships of TMEM16A, it is necessary to fully clarify the structural basis of the gating of the TMEM16A channel. Herein, we performed the protein electrostatic analysis and molecular dynamics simulation on the TMEM16A in the presence and absence of Ca2+. Data showed that the separation of TM4 and TM6 causes pore expansion, and Q646 may be a key residue for the formation of π-helix in the middle segment of TM6. Moreover, E705 was found to form a group of H-bond interactions with D554/K588/K645 below the hydrophobic gate to stabilize the closed conformation of the pore in the Ca2+-free state. Interestingly, in the Ca2+ bound state, the E705 side chain swings 100o to serve as Ca2+-binding coordination and released K645. K645 is closer to the hydrophobic gate in the calcium-bound state, which facilitates the provision of electrostatic forces for chloride ions as the ions pass through the hydrophobic gate. Our findings provide the structural-based insights to understanding the mechanisms of gating of TMEM16A.


Asunto(s)
Anoctamina-1/ultraestructura , Comunicación Celular/genética , Conformación Proteica en Hélice alfa/genética , Conformación Proteica , Anoctamina-1/química , Anoctamina-1/genética , Calcio/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Electricidad Estática , Relación Estructura-Actividad
4.
Cells ; 10(11)2021 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-34831090

RESUMEN

Cystic fibrosis (CF) is the most common of rare hereditary diseases in Caucasians, and it is estimated to affect 75,000 patients globally. CF is a complex disease due to the multiplicity of mutations found in the CF transmembrane conductance regulator (CFTR) gene causing the CFTR protein to become dysfunctional. Correctors and potentiators have demonstrated good clinical outcomes for patients with specific gene mutations; however, there are still patients for whom those treatments are not suitable and require alternative CFTR-independent strategies. Although CFTR is the main chloride channel in the lungs, others could, e.g., anoctamin-1 (ANO1 or TMEM16A), compensate for the deficiency of CFTR. This review summarizes the current knowledge on calcium-activated chloride channel (CaCC) ANO1 and presents ANO1 as an exciting target in CF.


Asunto(s)
Anoctamina-1/metabolismo , Fibrosis Quística/tratamiento farmacológico , Animales , Anoctamina-1/química , Fibrosis Quística/patología , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Modelos Biológicos
5.
Protein Pept Lett ; 28(12): 1338-1348, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34749600

RESUMEN

TMEM16A mediates the calcium-activated transmembrane flow of chloride ions and a variety of physiological functions. The binding of cytoplasmic calcium ions of TMEM16A and the consequent conformational changes of it are the key issues to explore the structure-function relationship. In recent years, researchers have explored this issue through electrophysiological experiments, structure resolving, molecular dynamic simulation, and other methods. The structures of TMEM16 family members determined by cryo-Electron microscopy (cryo-EM) and X-ray crystallization provide the primary basis for the investigation of the molecular mechanism of TMEM16A. However, the binding and activation mechanism of calcium ions in TMEM16A are still unclear and controversial. This mini-review discusses four Ca2+ sensing sites of TMEM16A and analyzes activation properties of TMEM16A by them, which will help understand the structure-function relationship of TMEM16A and throw light on the molecular design targeting the TMEM16A channel.


Asunto(s)
Anoctamina-1/química , Calcio/química , Proteínas de Neoplasias/química , Animales , Anoctamina-1/metabolismo , Sitios de Unión , Calcio/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos , Proteínas de Neoplasias/metabolismo , Relación Estructura-Actividad
6.
Biochim Biophys Acta Mol Cell Res ; 1868(12): 119132, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34450215

RESUMEN

Anoctamin-1 (ANO1), also known as transmembrane protein 16A (TMEM16A), is identified as a Ca2+-activated Cl- channel that is expressed in many organs and tissues. It is involved in numerous major physiological functions and especially in tumor growth. By screening 530 natural compounds, we identified cepharanthine as a potent blocker of ANO1 channels with an IC50 of 11.2 ± 0.9 µM and Emax of 92.7 ± 1.7%. The Lys384, Arg535, Thr539, and Glu624 in ANO1 are critical for the inhibitory effect of cepharanthine. Similar to its effect on ANO1, cepharanthine inhibits ANO2, the closest analog of TMEM16A. In contrast, up to 30 µM of cepharanthine showed limited inhibitory effects on recombinant ANO6 and bestrophin-1-encoded Ca2+-activated Cl- currents, but it showed no effects on endogenous volume-regulated anion currents (VRAC). Cepharanthine could also potently suppress endogenous ANO1 currents, significantly inhibit cell proliferation and migration, and induce apoptosis in LA795 lung adenocarcinoma cells. Moreover, animal experiments have shown that cepharanthine can dramatically inhibit the growth of xenograft tumors in mice. The high specificity provided by cepharanthine could be an important foundation for future studies of the physiological role of ANO1 channels, and these findings may reveal a new mechanism of its anticancer effect.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Anoctamina-1/antagonistas & inhibidores , Antineoplásicos Fitogénicos/farmacología , Bencilisoquinolinas/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Anoctamina-1/química , Anoctamina-1/metabolismo , Antineoplásicos Fitogénicos/uso terapéutico , Bencilisoquinolinas/uso terapéutico , Sitios de Unión , Células CHO , Cricetinae , Cricetulus , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Unión Proteica
7.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920953

RESUMEN

TMEM16A is a Ca2+-activated Cl- channel that controls broad cellular processes ranging from mucus secretion to signal transduction and neuronal excitability. Recent studies have reported that membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is an important cofactor that allosterically regulates TMEM16A channel activity. However, the detailed regulatory actions of PIP2 in splice variants of TMEM16A remain unclear. Here, we demonstrated that the attenuation of membrane phosphoinositide levels selectively inhibited the current amplitude of the TMEM16A(ac) isoform by decreasing the slow, but not instantaneous, Cl- currents, which are independent of the membrane potential and specific to PI(4,5)P2 depletion. The attenuation of endogenous PI(4,5)P2 levels by the activation of Danio rerio voltage-sensitive phosphatase (Dr-VSP) decreased the Cl- currents of TMEM16A(ac) but not the TMEM16A(a) isoform, which was abolished by the co-expression of PIP 5-kinase type-1γ (PIPKIγ). Using the rapamycin-inducible dimerization of exogenous phosphoinositide phosphatases, we further revealed that the stimulatory effects of phosphoinositide on TMEM16A(ac) channels were similar in various membrane potentials and specific to PI(4,5)P2, not PI4P and PI(3,4,5)P3. Finally, we also confirmed that PI(4,5)P2 resynthesis is essential for TMEM16A(ac) recovery from Dr-VSP-induced current inhibition. Our data demonstrate that membrane PI(4,5)P2 selectively modulates the gating of the TMEM16A(ac) channel in an agonistic manner, which leads to the upregulation of TMEM16A(ac) functions in physiological conditions.


Asunto(s)
Empalme Alternativo/genética , Anoctamina-1/genética , Calcio/metabolismo , Membrana Celular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Empalme Alternativo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Anoctamina-1/química , Anoctamina-1/metabolismo , Membrana Celular/efectos de los fármacos , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Monoéster Fosfórico Hidrolasas/metabolismo , Receptor Muscarínico M1/metabolismo , Sirolimus/farmacología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
8.
Int J Mol Sci ; 22(4)2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33672260

RESUMEN

Intracellular divalent cations control the molecular function of transmembrane protein 16 (TMEM16) family members. Both anion channels (such as TMEM16A) and phospholipid scramblases (such as TMEM16F) in this family are activated by intracellular Ca2+ in the low µM range. In addition, intracellular Ca2+ or Co2+ at mM concentrations have been shown to further potentiate the saturated Ca2+-activated current of TMEM16A. In this study, we found that all alkaline earth divalent cations in mM concentrations can generate similar potentiation effects in TMEM16A when applied intracellularly, and that manipulations thought to deplete membrane phospholipids weaken the effect. In comparison, mM concentrations of divalent cations minimally potentiate the current of TMEM16F but significantly change its cation/anion selectivity. We suggest that divalent cations may increase local concentrations of permeant ions via a change in pore electrostatic potential, possibly acting through phospholipid head groups in or near the pore. Monovalent cations appear to exert a similar effect, although with a much lower affinity. Our findings resolve controversies regarding the ion selectivity of TMEM16 proteins. The physiological role of this mechanism, however, remains elusive because of the nearly constant high cation concentrations in cytosols.


Asunto(s)
Anoctaminas/metabolismo , Cationes Bivalentes/metabolismo , Anoctamina-1/química , Anoctamina-1/genética , Anoctamina-1/metabolismo , Anoctaminas/química , Anoctaminas/genética , Calcio/metabolismo , Cationes Bivalentes/farmacología , Cobalto/metabolismo , Electrofisiología/métodos , Células HEK293 , Humanos , Magnesio/metabolismo , Manitol/metabolismo , Manitol/farmacología , Mutación , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolípidos/metabolismo , Polilisina/farmacología
9.
Cell Rep ; 33(13): 108570, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33378669

RESUMEN

Calcium (Ca2+) is the primary stimulus for transmembrane protein 16 (TMEM16) Ca2+-activated chloride channels and phospholipid scramblases, which regulate important physiological processes ranging from smooth muscle contraction to blood coagulation and tumor progression. Binding of intracellular Ca2+ to two highly conserved orthosteric binding sites in transmembrane helices (TMs) 6-8 efficiently opens the permeation pathway formed by TMs 3-7. Recent structures of TMEM16K and TMEM16F scramblases revealed an additional Ca2+ binding site between TM2 and TM10, whose functional relevance remains unknown. Here, we report that Ca2+ binds with high affinity to the equivalent third Ca2+ site in TMEM16A to enhance channel activation. Our cadmium (Cd2+) metal bridging experiments reveal that the third Ca2+ site's conformational states can profoundly influence TMEM16A's opening. Our study thus confirms the existence of a third Ca2+ site in TMEM16A, defines its functional importance in channel gating, and provides insight into a long-range allosteric gating mechanism of TMEM16 channels and scramblases.


Asunto(s)
Anoctamina-1/fisiología , Calcio/metabolismo , Canales de Cloruro/fisiología , Anoctamina-1/química , Sitios de Unión , Cadmio/metabolismo , Membrana Celular/metabolismo , Electrofisiología/métodos , Células HEK293 , Humanos , Activación del Canal Iónico , Transporte Iónico , Modelos Moleculares , Mutación , Proteínas de Transferencia de Fosfolípidos/fisiología , Conformación Proteica , Dominios Proteicos
10.
Proc Natl Acad Sci U S A ; 117(48): 30787-30798, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33199590

RESUMEN

Transmembrane 16A (TMEM16A, anoctamin1), 1 of 10 TMEM16 family proteins, is a Cl- channel activated by intracellular Ca2+ and membrane voltage. This channel is also regulated by the membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. We find that two splice variants of TMEM16A show different sensitivity to endogenous PI(4,5)P2 degradation, where TMEM16A(ac) displays higher channel activity and more current inhibition by PI(4,5)P2 depletion than TMEM16A(a). These two channel isoforms differ in the alternative splicing of the c-segment (exon 13). The current amplitude and PI(4,5)P2 sensitivity of both TMEM16A(ac) and (a) are significantly strengthened by decreased free cytosolic ATP and by conditions that decrease phosphorylation by Ca2+/calmodulin-dependent protein kinase II (CaMKII). Noise analysis suggests that the augmentation of currents is due to a rise of single-channel current (i), but not of channel number (N) or open probability (PO). Mutagenesis points to arginine 486 in the first intracellular loop as a putative binding site for PI(4,5)P2, and to serine 673 in the third intracellular loop as a site for regulatory channel phosphorylation that modulates the action of PI(4,5)P2 In silico simulation suggests how phosphorylation of S673 allosterically and differently changes the structure of the distant PI(4,5)P2-binding site between channel splice variants with and without the c-segment exon. In sum, our study reveals the following: differential regulation of alternatively spliced TMEM16A(ac) and (a) by plasma membrane PI(4,5)P2, modification of these effects by channel phosphorylation, identification of the molecular sites, and mechanistic explanation by in silico simulation.


Asunto(s)
Empalme Alternativo , Anoctamina-1/genética , Anoctamina-1/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Fosfatidilinositoles/metabolismo , Regulación Alostérica , Animales , Anoctamina-1/química , Sitios de Unión , Membrana Celular/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Ratones , Modelos Moleculares , Conformación Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Isoformas de Proteínas , Relación Estructura-Actividad
11.
Arch Biochem Biophys ; 695: 108650, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33132191

RESUMEN

TMEM16A is a calcium-activated chloride channel that is associate with several diseases, including pulmonary diseases, hypertension, diarrhea and cancer. The CaCCinh-A01 (A01) is widely recognized as an efficient blocker of TMEM16A and has been used as a tool drug to inhibit TMEM16A currents in the laboratory. A01 also has excellent pharmacokinetic properties and can be developed as a drug to target TMEM16A. However, the molecular mechanism how A01 inhibits TMEM16A is still elusive, which slows down its drug development process. Here, calculations identified that the binding pocket of A01 was located above the pore, and it was also discovered that the binding of A01 to TMEM16A not only blocked the pore but also led to its collapse. The interaction model analysis predicted that R515/K603/E623 were crucial residues for the binding between TMEM16A and A01, and the site-directed mutagenesis studies confirmed the above results. The binding mode and quantum chemical calculations showed that the carboxyl and the amide oxygen atom of A01 were the key interaction sites between TMEM16A and A01. Therefore, our study proposed the inhibitory mechanism of TMEM16A current by A01 and revealed how A01 inhibits TMEM16A at the molecular level. These findings will shed light on both the development of A01 as a potential drug for TMEM16A dysfunction-related disorders and drug screening targeting the pocket.


Asunto(s)
Anoctamina-1 , Simulación del Acoplamiento Molecular , Proteínas de Neoplasias , Tiofenos/química , Sustitución de Aminoácidos , Anoctamina-1/antagonistas & inhibidores , Anoctamina-1/química , Anoctamina-1/genética , Anoctamina-1/metabolismo , Sitios de Unión , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Mutación Missense , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
12.
Cell Rep ; 30(4): 1141-1151.e3, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995732

RESUMEN

The secreted protein calcium-activated chloride channel regulator 1 (CLCA1) utilizes a von Willebrand factor type A (VWA) domain to bind to and potentiate the calcium-activated chloride channel TMEM16A. To gain insight into this unique potentiation mechanism, we determined the 2.0-Å crystal structure of human CLCA1 VWA bound to Ca2+. The structure reveals the metal-ion-dependent adhesion site (MIDAS) in a high-affinity "open" conformation, engaging in crystal contacts that likely mimic how CLCA1 engages TMEM16A. The CLCA1 VWA contains a disulfide bond between α3 and α4 in close proximity to the MIDAS that is invariant in the CLCA family and unique in VWA structures. Further biophysical studies indicate that CLCA1 VWA is preferably stabilized by Mg2+ over Ca2+ and that α6 atypically extends from the VWA core. Finally, an analysis of TMEM16A structures suggests residues likely to mediate interaction with CLCA1 VWA.


Asunto(s)
Anoctamina-1/química , Anoctamina-1/metabolismo , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Fenómenos Biofísicos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Dominios Proteicos , Pliegue de Proteína
13.
Biophys J ; 118(1): 262-272, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31818463

RESUMEN

The calcium-activated chloride channel TMEM16A is involved in many physiological processes, and insufficient function of TMEM16A may lead to the occurrence of various diseases. Therefore, TMEM16A activators are supposed to be potentially useful for treatment of TMEM16A downregulation-inducing diseases. However, the TMEM16A activators are relatively rare, and the underlying activation mechanism of them is unclear. In the previous work, we have proved that ginsenoside Rb1 is a TMEM16A activator. In this work, we explored the activation mechanism of ginsenoside analogs on TMEM16A through analyzing the interactions between six ginsenoside analogs and TMEM16A. We identified GRg2 and GRf can directly activate TMEM16A by screening five novel ginsenosids analogs (GRb2, GRf, GRg2, GRh2, and NGR1). Isolated guinea pig ileum assay showed both GRg2 and GRf increased the amplitude and frequency of ileum contractions. We explored the molecular mechanisms of ginsenosides activating TMEM16A by combining molecular simulation with electrophysiological experiments. We proposed a TMEM16A activation process model based on the results, in which A697 on TM7 and L746 on TM8 bind to the isobutenyl of ginsenosides through hydrophobic interaction to fix the spatial location of ginsenosides. N650 on TM6 and E705 on TM7 bind to ginsenosides through electrostatic interaction, which causes the inner half of α-helix 6 to form physical contact with ginsenosides and leads to the pore opening. It should be emphasized that TMEM16A can be activated by ginsenosides only when both the above two conditions are satisfied. This is the first, to our knowledge, report of TMEM16A opening process activated by small-molecule activators. The mechanism of ginsenosides activating TMEM16A will provide important clues for TMEM16A gating mechanism and for new TMEM16A activators screening.


Asunto(s)
Anoctamina-1/metabolismo , Ginsenósidos/química , Ginsenósidos/farmacología , Animales , Anoctamina-1/química , Sitios de Unión , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Ginsenósidos/metabolismo , Cobayas , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformación Proteica , Electricidad Estática
14.
Proc Natl Acad Sci U S A ; 116(40): 19952-19962, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31515451

RESUMEN

ANO1 (TMEM16A) is a Ca2+-activated Cl- channel that regulates diverse cellular functions including fluid secretion, neuronal excitability, and smooth muscle contraction. ANO1 is activated by elevation of cytosolic Ca2+ and modulated by phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Here, we describe a closely concerted experimental and computational study, including electrophysiology, mutagenesis, functional assays, and extended sampling of lipid-protein interactions with molecular dynamics (MD) to characterize PI(4,5)P2 binding modes and sites on ANO1. ANO1 currents in excised inside-out patches activated by 270 nM Ca2+ at +100 mV are increased by exogenous PI(4,5)P2 with an EC50 = 1.24 µM. The effect of PI(4,5)P2 is dependent on membrane voltage and Ca2+ and is explained by a stabilization of the ANO1 Ca2+-bound open state. Unbiased atomistic MD simulations with 1.4 mol% PI(4,5)P2 in a phosphatidylcholine bilayer identified 8 binding sites with significant probability of binding PI(4,5)P2 Three of these sites captured 85% of all ANO1-PI(4,5)P2 interactions. Mutagenesis of basic amino acids near the membrane-cytosol interface found 3 regions of ANO1 critical for PI(4,5)P2 regulation that correspond to the same 3 sites identified by MD. PI(4,5)P2 is stabilized by hydrogen bonding between amino acid side chains and phosphate/hydroxyl groups on PI(4,5)P2 Binding of PI(4,5)P2 alters the position of the cytoplasmic extension of TM6, which plays a crucial role in ANO1 channel gating, and increases the accessibility of the inner vestibule to Cl- ions. We propose a model consisting of a network of 3 PI(4,5)P2 binding sites at the cytoplasmic face of the membrane allosterically regulating ANO1 channel gating.


Asunto(s)
Anoctamina-1/química , Calcio/química , Proteínas de Neoplasias/química , Fosfatidilinositol 4,5-Difosfato/química , Sitios de Unión , Cationes , Citosol/metabolismo , Electrofisiología , Células HEK293 , Humanos , Conformación Molecular , Simulación de Dinámica Molecular , Mutagénesis , Mutación , Técnicas de Placa-Clamp , Fosfolípidos/química , Relación Estructura-Actividad
15.
Int J Biochem Cell Biol ; 111: 37-41, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31005634

RESUMEN

The molecular identity of calcium-activated chloride channels (CaCCs) was clarified only some ten years ago when it was linked to the family of "transmembrane proteins of unknown function 16″ (TMEM16). Since then, numerous studies have been conducted both to define their role in physiology and identify their biophysical functions. For the latter, the ultrastructural description of mouse TMEM16 A was a breakthrough. CaCCs were functionally described in a number of different tissues including first-order sensory neurons. The activating rise in intracellular calcium concentration can be caused by an influx of calcium through other calcium permeable ion channels. Calcium release from intracellular stores, mediated by G-protein coupled receptors, also leads to CaCC activation. Prominent inflammatory mediators like bradykinin or serotonin stimulate CaCCs via such a mechanism. The (patho) physiological function of these ion channels renders them promising targets for antinociceptive treatment.


Asunto(s)
Analgésicos/farmacología , Canales de Cloruro/metabolismo , Terapia Molecular Dirigida/métodos , Animales , Anoctamina-1/química , Anoctamina-1/metabolismo , Canales de Cloruro/química , Humanos , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo
16.
J Gen Physiol ; 151(4): 518-531, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30670476

RESUMEN

Two TMEM16 family members, TMEM16A and TMEM16F, have different ion transport properties. Upon activation by intracellular Ca2+, TMEM16A-a Ca2+-activated Cl- channel-is more selective for anions than cations, whereas TMEM16F-a phospholipid scramblase-appears to transport both cations and anions. Under saturating Ca2+ conditions, the current-voltage (I-V) relationships of these two proteins also differ; the I-V curve of TMEM16A is linear, while that of TMEM16F is outwardly rectifying. We previously found that mutating a positively charged lysine residue (K584) in the ion transport pathway to glutamine converted the linear I-V curve of TMEM16A to an outwardly rectifying curve. Interestingly, the corresponding residue in the outwardly rectifying TMEM16F is also a glutamine (Q559). Here, we examine the ion transport functions of TMEM16 molecules and compare the roles of K584 of TMEM16A and Q559 of TMEM16F in controlling the rectification of their respective I-V curves. We find that rectification of TMEM16A is regulated electrostatically by the side-chain charge on the residue at position 584, whereas the charge on residue 559 in TMEM16F has little effect. Unexpectedly, mutation of Q559 to aromatic amino acid residues significantly alters outward rectification in TMEM16F. These same mutants show reduced Ca2+-induced current rundown (or desensitization) compared with wild-type TMEM16F. A mutant that removes the rundown of TMEM16F could facilitate the study of ion transport mechanisms in this phospholipid scramblase in the same way that a CLC-0 mutant in which inactivation (or closure of the slow gate) is suppressed was used in our previous studies.


Asunto(s)
Anoctamina-1/química , Anoctamina-1/fisiología , Proteínas de Transferencia de Fosfolípidos/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Fenómenos Electrofisiológicos , Transporte Iónico , Ratones , Mutación , Proteínas de Transferencia de Fosfolípidos/genética , Isoformas de Proteínas
17.
Elife ; 72018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30311910

RESUMEN

TMEM16A is a ligand-gated anion channel that is activated by intracellular Ca2+. This channel comprises two independent pores and closely apposed Ca2+ binding sites that are contained within each subunit of a homodimeric protein. Previously we characterized the influence of positively charged pore-lining residues on anion conduction (Paulino et al., 2017a). Here, we demonstrate the electrostatic control of permeation by the bound calcium ions in mouse TMEM16A using electrophysiology and Poisson-Boltzmann calculations. The currents of constitutively active mutants lose their outward rectification as a function of Ca2+ concentration due to the alleviation of energy barriers for anion conduction. This phenomenon originates from Coulombic interactions between the bound Ca2+ and permeating anions and thus demonstrates that an electrostatic gate imposed by the vacant binding site present in the sterically open pore, is released by Ca2+ binding to enable an otherwise sub-conductive pore to conduct with full capacity.


Asunto(s)
Anoctamina-1/genética , Calcio/metabolismo , Proteínas Mutantes/genética , Conformación Proteica , Animales , Aniones/química , Anoctamina-1/química , Sitios de Unión , Calcio/química , Agonistas de los Canales de Calcio , Células HEK293 , Humanos , Activación del Canal Iónico/genética , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Electricidad Estática
18.
Biochem Biophys Res Commun ; 503(3): 1710-1715, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30078682

RESUMEN

Anoctamin 1 (encoded by the Ano1 gene) is a Ca2+-activated Cl- channel critical to many physiological functions. It has been speculated that Ano1 expression is regulated in a tissue-dependent manner via alternative promoters. However, variation in the 5'-end sequence of mouse Ano1 (mAno1) and its tissue-dependent regulation are poorly understood. We identified a novel 5'-terminal exon (designated exon 1a) of mAno1 instead of the known 5'-terminal exon (exon 0) using 5'-rapid amplification of cDNA ends (RACE) analysis. Unexpectedly, the novel 5'-end variant mAno1Ex1a was abundantly expressed in many tissues including the salivary and mammary glands, rectum, lung, trachea and prostate. In contrast, the known variant mAno1Ex0 predominated only in male reproductive tissues such as the epididymis and testis. In a heterologous expression system, mAno1Ex0 encoded a longer protein than mAno1Ex1a, and this long isoform was abolished by a mutation in the exon 0 start codon. Moreover, the mAno1Ex0-specific N-terminal sequence was immunohistochemically detected in epididymis but not in salivary gland. Our data suggest that mAno1 expression is regulated via alternative promoters, and its transcriptional variation results in variation of the N-terminal sequence of the Ano1 protein due to the alternative translation initiation sites. These tissue-specific variations might contribute to the regulation of mAno1 expression and activity according to the physiological function of each tissue.


Asunto(s)
Anoctamina-1/química , Anoctamina-1/genética , Exones/genética , Variación Genética/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Animales , Anoctamina-1/análisis , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética
19.
Int J Mol Sci ; 19(5)2018 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-29748496

RESUMEN

Calcium-activated chloride secretion in epithelial tissues has been described for many years. However, the molecular identity of the channel responsible for the Ca2+-activated Cl− secretion in epithelial tissues has remained a mystery. More recently, TMEM16A has been identified as a new putative Ca2+-activated Cl− channel (CaCC). The primary goal of this article will be to review the characterization of TMEM16A, as it relates to the physical structure of the channel, as well as important residues that confer voltage and Ca2+-sensitivity of the channel. This review will also discuss the role of TMEM16A in epithelial physiology and potential associated-pathophysiology. This will include discussion of developed knockout models that have provided much needed insight on the functional localization of TMEM16A in several epithelial tissues. Finally, this review will examine the implications of the identification of TMEM16A as it pertains to potential novel therapies in several pathologies.


Asunto(s)
Anoctamina-1/genética , Señalización del Calcio/genética , Canales de Cloruro/genética , Proteínas de Neoplasias/genética , Anoctamina-1/química , Calcio/química , Agonistas de los Canales de Calcio/química , Canales de Cloruro/química , Cloruros/química , Epitelio/química , Epitelio/metabolismo , Humanos , Proteínas de Neoplasias/química
20.
Neuron ; 97(5): 1063-1077.e4, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29478917

RESUMEN

Calcium-activated chloride channels (CaCCs) formed by TMEM16A or TMEM16B are broadly expressed in the nervous system, smooth muscles, exocrine glands, and other tissues. With two calcium-binding sites and a pore within each monomer, the dimeric CaCC exhibits voltage-dependent calcium sensitivity. Channel activity also depends on the identity of permeant anions. To understand how CaCC regulates neuronal signaling and how CaCC is, in turn, modulated by neuronal activity, we examined the molecular basis of CaCC gating. Here, we report that voltage modulation of TMEM16A-CaCC involves voltage-dependent occupancy of calcium- and anion-binding site(s) within the membrane electric field as well as a voltage-dependent conformational change intrinsic to the channel protein. These gating modalities all critically depend on the sixth transmembrane segment.


Asunto(s)
Anoctamina-1/química , Anoctamina-1/metabolismo , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Activación del Canal Iónico/fisiología , Secuencia de Aminoácidos , Animales , Anoctamina-1/genética , Canales de Cloruro/genética , Células HEK293 , Humanos , Ratones , Unión Proteica/fisiología , Estructura Secundaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...