Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39071375

RESUMEN

Assembly of NADPH oxidase 2 (NOX2) proteins in neutrophils plays an essential role in controlling microbial infections by producing high levels of reactive oxygen species (ROS). In contrast, the role of the Hv1 voltage-gated proton channel that is required for sustained NOX2 activity is less well characterized. We examined the role of Hv1 in a murine model of blinding Pseudomonas aeruginosa corneal infection and found that in contrast to C57BL/6 mice, Hvcn1 -/- mice exhibit an impaired ability to kill bacteria and regulate disease severity. In vitro, we used a novel Hv1 Inhibitor Flexible (HIF) to block ROS production by human and murine neutrophils and found that HIF inhibits ROS production in a dose-dependent manner following stimulation with PMA or infection with P. aeruginosa. Collectively, these findings demonstrate an important role for Hv1 on controlling bacterial growth in a clinically relevant bacterial infection model.

2.
Nat Commun ; 14(1): 7515, 2023 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-37980353

RESUMEN

Voltage-gated and mechanically-gated ion channels are distinct classes of membrane proteins that conduct ions across gated pores and are turned on by electrical or mechanical stimuli, respectively. Here, we describe an Hv channel (a.k.a voltage-dependent H+ channel) from the angiosperm plant A. thaliana that gates with a unique modality as it is turned on by an electrical stimulus only after exposure to a mechanical stimulus, a process that we call priming. The channel localizes in the vascular tissue and has homologs in vascular plants. We find that mechanical priming is not required for activation of non-angiosperm Hvs. Guided by AI-generated structural models of plant Hv homologs, we identify a set of residues playing a crucial role in mechanical priming. We propose that Hvs from angiosperm plants require priming because of a network of hydrophilic/charged residues that locks the channels in a silent resting conformation. Mechanical stimuli destabilize the network allowing the conduction pathway to turn on. In contrast to many other channels and receptors, Hv proteins are not thought to possess mechanisms such as inactivation or desensitization. Our findings demonstrate that angiosperm Hv channels are electrically silent until a mechanical stimulation turns on their voltage-dependent activity.


Asunto(s)
Magnoliopsida , Tracheophyta , Protones , Magnoliopsida/metabolismo , Canales Iónicos/metabolismo , Tracheophyta/metabolismo
3.
Nanotechnology ; 34(46)2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37524056

RESUMEN

We describe the concept and roadmap of an engineered electronic nose with specificity towards analytes that differ by as little as one carbon atom, and sensitivity of being able to electrically register a single molecule of analyte. The analyte could be anything that natural noses can detect, e.g. trinitrotoluene (TNT), cocaine, aromatics, volatile organic compounds etc. The strategy envisioned is to genetically engineer a fused olfactory odorant receptor (odorant receptor (OR), a membrane-bound G-protein coupled receptor (GPCR) with high selectivity) to an ion channel protein, which opens in response to binding of the ligand to the OR. The lipid bilayer supporting the fused sensing protein would be intimately attached to a nanowire or nanotube network (either via a covalent tether or a non-covalent physisorption process), which would electrically detect the opening of the ion channel, and hence the binding of a single ligand to a single OR protein domain. Three man-made technological advances: (1) fused GPCR to ion channel protein, (2) nanowire sensing of single ion channel activity, and (3) lipid bilayer to nanotube/nanowire tethering chemistry and on natural technology (sensitivity and selectivity of OR domains to specific analytes) each have been demonstrated and/or studied independently. The combination of these three technological advances and the result of millions of years of evolution of OR proteins would enable the goal of single molecule sensing with specificity towards analytes that differ by as little as one carbon atom. This is both a review of the past and a vision of the future.


Asunto(s)
Técnicas Biosensibles , Nanocables , Receptores Odorantes , Humanos , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Membrana Dobles de Lípidos , Nariz Electrónica , Ligandos , Canales Iónicos
4.
J Gen Physiol ; 153(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34228044

RESUMEN

The human voltage-gated proton channel Hv1 is a drug target for cancer, ischemic stroke, and neuroinflammation. It resides on the plasma membrane and endocytic compartments of a variety of cell types, where it mediates outward proton movement and regulates the activity of NOX enzymes. Its voltage-sensing domain (VSD) contains a gated and proton-selective conduction pathway, which can be blocked by aromatic guanidine derivatives such as 2-guanidinobenzimidazole (2GBI). Mutation of Hv1 residue F150 to alanine (F150A) was previously found to increase 2GBI apparent binding affinity more than two orders of magnitude. Here, we explore the contribution of aromatic interactions between the inhibitor and the channel in the presence and absence of the F150A mutation, using a combination of electrophysiological recordings, classic mutagenesis, and site-specific incorporation of fluorinated phenylalanines via nonsense suppression methodology. Our data suggest that the increase in apparent binding affinity is due to a rearrangement of the binding site allowed by the smaller residue at position 150. We used this information to design new arginine mimics with improved affinity for the nonrearranged binding site of the wild-type channel. The new compounds, named "Hv1 Inhibitor Flexibles" (HIFs), consist of two "prongs," an aminoimidazole ring, and an aromatic group connected by extended flexible linkers. Some HIF compounds display inhibitory properties that are superior to those of 2GBI, thus providing a promising scaffold for further development of high-affinity Hv1 inhibitors.


Asunto(s)
Arginina , Canales Iónicos , Sitios de Unión , Humanos , Canales Iónicos/metabolismo , Ligandos , Protones
5.
J Gen Physiol ; 153(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34228045

RESUMEN

Voltage-gated sodium, potassium, and calcium channels consist of four voltage-sensing domains (VSDs) that surround a central pore domain and transition from a down state to an up state in response to membrane depolarization. While many types of drugs bind pore domains, the number of organic molecules known to bind VSDs is limited. The Hv1 voltage-gated proton channel is made of two VSDs and does not contain a pore domain, providing a simplified model for studying how small ligands interact with VSDs. Here, we describe a ligand, named HIF, that interacts with the Hv1 VSD in the up and down states. We find that HIF rapidly inhibits proton conduction in the up state by blocking the open channel, as previously described for 2-guanidinobenzimidazole and its derivatives. HIF, however, interacts with a site slowly accessible in the down state. Functional studies and MD simulations suggest that this interaction traps the compound in a narrow pocket lined with charged residues within the VSD intracellular vestibule, which results in slow recovery from inhibition. Our findings point to a "wrench in gears" mechanism whereby side chains within the binding pocket trap the compound as the teeth of interlocking gears. We propose that the use of screening strategies designed to target binding sites with slow accessibility, similar to the one identified here, could lead to the discovery of new ligands capable of interacting with VSDs of other voltage-gated ion channels in the down state.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos , Canales Iónicos/metabolismo , Potasio , Protones
6.
Commun Biol ; 4(1): 261, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637875

RESUMEN

Here, we report the identification and characterization of the first proton channels from fungi. The fungal proteins are related to animal voltage-gated Hv channels and are conserved in both higher and lower fungi. Channels from Basidiomycota and Ascomycota appear to be evolutionally and functionally distinct. Representatives from the two phyla share several features with their animal counterparts, including structural organization and strong proton selectivity, but they differ from each other and from animal Hvs in terms of voltage range of activation, pharmacology, and pH sensitivity. The activation gate of Hv channels is believed to be contained within the transmembrane core of the protein and little is known about contributions of peripheral regions to the activation mechanism. Using a chimeragenesis approach, we find that intra- and extracellular peripheral regions are main determinants of the voltage range of activation in fungal channels, highlighting the role of these overlooked components in channel gating.


Asunto(s)
Ascomicetos/metabolismo , Basidiomycota/metabolismo , Proteínas Fúngicas/metabolismo , Activación del Canal Iónico , Canales Iónicos/metabolismo , Animales , Antifúngicos/farmacología , Ascomicetos/efectos de los fármacos , Ascomicetos/genética , Basidiomycota/efectos de los fármacos , Basidiomycota/genética , Evolución Molecular , Proteínas Fúngicas/antagonistas & inhibidores , Proteínas Fúngicas/genética , Concentración de Iones de Hidrógeno , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Mecanotransducción Celular , Potenciales de la Membrana , Dominios y Motivos de Interacción de Proteínas , Protones , Xenopus
7.
J Membr Biol ; 254(1): 5-16, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33196887

RESUMEN

The voltage-gated proton channel Hv1 mediates efflux of protons from the cell. Hv1 integrally contributes to various physiological processes including pH homeostasis and the respiratory burst of phagocytes. Inhibition of Hv1 may provide therapeutic avenues for the treatment of inflammatory diseases, breast cancer, and ischemic brain damage. In this work, we investigate two prototypical Hv1 inhibitors, 2-guanidinobenzimidazole (2GBI), and 5-chloro-2-guanidinobenzimidazole (GBIC), from an experimentally screened class of guanidine derivatives. Both compounds block proton conduction by binding the same site located on the intracellular side of the channel. However, when added to the extracellular medium, the compounds strongly differ in their ability to inhibit proton conduction, suggesting substantial differences in membrane permeability. Here, we compute the potential of mean force for each compound to permeate through the membrane using atomistic molecular dynamics simulations with the adaptive biasing force method. Our results rationalize the putative distinction between these two blockers with respect to their abilities to permeate the cellular membrane.


Asunto(s)
Canales Iónicos/antagonistas & inhibidores , Termodinámica , Permeabilidad de la Membrana Celular , Canales Iónicos/metabolismo , Simulación de Dinámica Molecular , Protones
8.
Sci Rep ; 10(1): 13587, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788614

RESUMEN

Hv1 is a voltage-gated proton channel whose main function is to facilitate extrusion of protons from the cell. The development of effective channel blockers for Hv1 can lead to new therapeutics for the treatment of maladies related to Hv1 dysfunction. Although the mechanism of proton permeation in Hv1 remains to be elucidated, a series of small molecules have been discovered to inhibit Hv1. Here, we computed relative binding free energies of a prototypical Hv1 blocker on a model of human Hv1 in an open state. We used alchemical free energy perturbation techniques based on atomistic molecular dynamics simulations. The results support our proposed open state model and shed light on the preferred tautomeric state of the channel blocker. This work lays the groundwork for future studies on adapting the blocker molecule for more effective inhibition of Hv1.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Iónicos/metabolismo , Simulación de Dinámica Molecular , Protones , Bibliotecas de Moléculas Pequeñas/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/química , Estructura Molecular , Unión Proteica , Conformación Proteica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Termodinámica
9.
Proc Natl Acad Sci U S A ; 117(24): 13490-13498, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32461356

RESUMEN

The voltage-gated Hv1 proton channel is a ubiquitous membrane protein that has roles in a variety of cellular processes, including proton extrusion, pH regulation, production of reactive oxygen species, proliferation of cancer cells, and increased brain damage during ischemic stroke. A crystal structure of an Hv1 construct in a putative closed state has been reported, and structural models for the channel open state have been proposed, but a complete characterization of the Hv1 conformational dynamics under an applied membrane potential has been elusive. We report structural models of the Hv1 voltage-sensing domain (VSD), both in a hyperpolarized state and a depolarized state resulting from voltage-dependent conformational changes during a 10-µs-timescale atomistic molecular dynamics simulation in an explicit membrane environment. In response to a depolarizing membrane potential, the S4 helix undergoes an outward displacement, leading to changes in the VSD internal salt-bridge network, resulting in a reshaping of the permeation pathway and a significant increase in hydrogen bond connectivity throughout the channel. The total gating charge displacement associated with this transition is consistent with experimental estimates. Molecular docking calculations confirm the proposed mechanism for the inhibitory action of 2-guanidinobenzimidazole (2GBI) derived from electrophysiological measurements and mutagenesis. The depolarized structural model is also consistent with the formation of a metal bridge between residues located in the core of the VSD. Taken together, our results suggest that these structural models are representative of the closed and open states of the Hv1 channel.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos/química , Canales Iónicos/metabolismo , Cristalografía por Rayos X , Guanidinas/metabolismo , Humanos , Enlace de Hidrógeno , Canales Iónicos/genética , Potenciales de la Membrana , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Conformación Proteica , Protones
10.
ACS Biomater Sci Eng ; 6(3): 1311-1320, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33455403

RESUMEN

Stem cells have attracted significant attention due to their regenerative capabilities and their potential for the treatment of disease. Consequently, significant research effort has focused on the development of protein- and polypeptide-based materials as stem cell substrates and scaffolds. Here, we explore the ability of reflectin, a cephalopod structural protein, to support the growth of murine neural stem/progenitor cells (mNSPCs). We observe that the binding, growth, and differentiation of mNSPCs on reflectin films is comparable to that on more established protein-based materials. Moreover, we find that heparin selectively inhibits the adhesion of mNSPCs on reflectin, affording spatial control of cell growth and leading to a >30-fold change in cell density on patterned substrates. The described findings highlight the potential utility of reflectin as a stem cell culture material.


Asunto(s)
Cefalópodos , Células-Madre Neurales , Animales , Diferenciación Celular , Proliferación Celular , Ratones , Proteínas
11.
Commun Biol ; 2: 298, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396578

RESUMEN

Piezo channels transduce mechanical stimuli into electrical and chemical signals to powerfully influence development, tissue homeostasis, and regeneration. Studies on Piezo1 have largely focused on transduction of "outside-in" mechanical forces, and its response to internal, cell-generated forces remains poorly understood. Here, using measurements of endogenous Piezo1 activity and traction forces in native cellular conditions, we show that cellular traction forces generate spatially-restricted Piezo1-mediated Ca2+ flickers in the absence of externally-applied mechanical forces. Although Piezo1 channels diffuse readily in the plasma membrane and are widely distributed across the cell, their flicker activity is enriched near force-producing adhesions. The mechanical force that activates Piezo1 arises from Myosin II phosphorylation by Myosin Light Chain Kinase. We propose that Piezo1 Ca2+ flickers allow spatial segregation of mechanotransduction events, and that mobility allows Piezo1 channels to explore a large number of mechanical microdomains and thus respond to a greater diversity of mechanical cues.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Fibroblastos/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular , Miosina Tipo II/metabolismo , Células-Madre Neurales/metabolismo , Animales , Células Cultivadas , Humanos , Canales Iónicos/deficiencia , Canales Iónicos/genética , Masculino , Ratones Noqueados , Factores de Tiempo
12.
J Phys Chem B ; 121(15): 3804-3812, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28074656

RESUMEN

Voltage-sensing domains (VSDs) sense changes in the membrane electrostatic potential and, through conformational changes, regulate a specific function. The VSDs of wild-type voltage-dependent K+, Na+, and Ca2+ channels do not conduct ions, but they can become ion-permeable through pathological mutations in the VSD. Relatively little is known about the underlying mechanisms of conduction through VSDs. The most detailed studies have been performed on Shaker K+ channel variants in which ion conduction through the VSD is manifested in electrophysiology experiments as a voltage-dependent inward current, the so-called omega current, which appears when the VSDs are in their resting state conformation. Only monovalent cations appear to permeate the Shaker VSD via a pathway that is believed to be, at least in part, the same as that followed by the S4 basic side chains during voltage-dependent activation. We performed µs-time scale atomistic molecular dynamics simulations of a cation-conducting variant of the Shaker VSD under applied electric fields in an experimentally validated resting-state conformation, embedded in a lipid bilayer surrounded by solutions containing guanidinium chloride or potassium chloride. Our simulations provide insights into the Shaker VSD permeation pathway, the protein-ion interactions that control permeation kinetics, and the mechanism of voltage-dependent activation of voltage-gated ion channels.


Asunto(s)
Simulación de Dinámica Molecular , Canales de Potasio de la Superfamilia Shaker/química , Conductividad Eléctrica , Dominios Proteicos
13.
J Gen Physiol ; 148(5): 405-418, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27799320

RESUMEN

The voltage-gated proton channel, Hv1, is expressed in tissues throughout the body and plays important roles in pH homeostasis and regulation of NADPH oxidase. Hv1 operates in membrane compartments that experience strong mechanical forces under physiological or pathological conditions. In microglia, for example, Hv1 activity is potentiated by cell swelling and causes an increase in brain damage after stroke. The channel complex consists of two proton-permeable voltage-sensing domains (VSDs) linked by a cytoplasmic coiled-coil domain. Here, we report that these VSDs directly respond to mechanical stimuli. We find that membrane stretch facilitates Hv1 channel opening by increasing the rate of activation and shifting the steady-state activation curve to less depolarized potentials. In the presence of a transmembrane pH gradient, membrane stretch alone opens the channel without the need for strong depolarizations. The effect of membrane stretch persists for several minutes after the mechanical stimulus is turned off, suggesting that the channel switches to a "facilitated" mode in which opening occurs more readily and then slowly reverts to the normal mode observed in the absence of membrane stretch. Conductance simulations with a six-state model recapitulate all the features of the channel's response to mechanical stimulation. Hv1 mechanosensitivity thus provides a mechanistic link between channel activation in microglia and brain damage after stroke.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos/metabolismo , Animales , Membrana Celular/metabolismo , Humanos , Canales Iónicos/química , Potenciales de la Membrana , Dominios Proteicos , Estrés Mecánico , Xenopus
15.
ACS Appl Mater Interfaces ; 8(1): 278-84, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26703760

RESUMEN

Cephalopods possess remarkable camouflage capabilities, which are enabled by their complex skin structure and sophisticated nervous system. Such unique characteristics have in turn inspired the design of novel functional materials and devices. Within this context, recent studies have focused on investigating the self-assembly, optical, and electrical properties of reflectin, a protein that plays a key role in cephalopod structural coloration. Herein, we report the discovery that reflectin constitutes an effective material for the growth of human neural stem/progenitor cells. Our findings may hold relevance both for understanding cephalopod embryogenesis and for developing improved protein-based bioelectronic devices.


Asunto(s)
Materiales Biocompatibles/farmacología , Células-Madre Neurales/citología , Proteínas/farmacología , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Decapodiformes/química , Humanos , Microscopía Fluorescente , Células-Madre Neurales/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos
16.
Sci Rep ; 5: 14077, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26365828

RESUMEN

The Hv1 voltage-gated proton channel is a dimeric complex consisting of two voltage-sensing domains (VSDs), each containing a gated proton permeation pathway. Dimerization is controlled by a cytoplasmic coiled-coil domain. The transitions from the closed to the open state in the two VSDs are known to occur cooperatively; however, the underlying mechanism is poorly understood. Intersubunit interfaces play a critical role in allosteric processes; but, such interfaces have not been determined in the open Hv1 channel. Here we show that 2-guanidinothiazole derivatives block the two Hv1 VSDs in a cooperative way, and use one of the compounds as a probe of allosteric coupling between open subunits. We find that the extracellular ends of the first transmembrane segments of the VSDs form the intersubunit interface that mediates coupling between binding sites, while the coiled-coil domain does not directly participate in the process. We also find strong evidence that the channel's proton selectivity filter controls blocker binding cooperativity.


Asunto(s)
Canales Iónicos/antagonistas & inhibidores , Tiazoles/química , Regulación Alostérica , Sitios de Unión , Dimerización , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Mutagénesis Sitio-Dirigida , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Tiazoles/metabolismo
17.
Sci Rep ; 5: 8499, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25686615

RESUMEN

Neural stem and progenitor cell (NSPC) fate is strongly influenced by mechanotransduction as modulation of substrate stiffness affects lineage choice. Other types of mechanical stimuli, such as stretch (tensile strain), occur during CNS development and trauma, but their consequences for NSPC differentiation have not been reported. We delivered a 10% static equibiaxial stretch to NSPCs and examined effects on differentiation. We found static stretch specifically impacts NSPC differentiation into oligodendrocytes, but not neurons or astrocytes, and this effect is dependent on particular extracellular matrix (ECM)-integrin linkages. Generation of oligodendrocytes from NSPCs was reduced on laminin, an outcome likely mediated by the α6 laminin-binding integrin, whereas similar effects were not observed for NSPCs on fibronectin. Our data demonstrate a direct role for tensile strain in dictating the lineage choice of NSPCs and indicate the dependence of this phenomenon on specific substrate materials, which should be taken into account for the design of biomaterials for NSPC transplantation.


Asunto(s)
Diferenciación Celular , Matriz Extracelular , Células-Madre Neurales/citología , Estrés Mecánico , Animales , Células Cultivadas , Integrinas/metabolismo , Laminina/metabolismo , Ratones , Oligodendroglía/citología , Unión Proteica
18.
Proc Natl Acad Sci U S A ; 111(45): 16148-53, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349416

RESUMEN

Neural stem cells are multipotent cells with the ability to differentiate into neurons, astrocytes, and oligodendrocytes. Lineage specification is strongly sensitive to the mechanical properties of the cellular environment. However, molecular pathways transducing matrix mechanical cues to intracellular signaling pathways linked to lineage specification remain unclear. We found that the mechanically gated ion channel Piezo1 is expressed by brain-derived human neural stem/progenitor cells and is responsible for a mechanically induced ionic current. Piezo1 activity triggered by traction forces elicited influx of Ca(2+), a known modulator of differentiation, in a substrate-stiffness-dependent manner. Inhibition of channel activity by the pharmacological inhibitor GsMTx-4 or by siRNA-mediated Piezo1 knockdown suppressed neurogenesis and enhanced astrogenesis. Piezo1 knockdown also reduced the nuclear localization of the mechanoreactive transcriptional coactivator Yes-associated protein. We propose that the mechanically gated ion channel Piezo1 is an important determinant of mechanosensitive lineage choice in neural stem cells and may play similar roles in other multipotent stem cells.


Asunto(s)
Señalización del Calcio/fisiología , Activación del Canal Iónico/fisiología , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Células Madre Multipotentes/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Canales Iónicos/genética , Masculino , Células Madre Multipotentes/citología , Células-Madre Neurales/citología
19.
PLoS One ; 9(8): e105926, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25165868

RESUMEN

The Hv1 channel and voltage-sensitive phosphatases share with voltage-gated sodium, potassium, and calcium channels the ability to detect changes in membrane potential through voltage-sensing domains (VSDs). However, they lack the pore domain typical of these other channels. NaV, KV, and CaV proteins can be found in neurons and muscles, where they play important roles in electrical excitability. In contrast, VSD-containing proteins lacking a pore domain are found in non-excitable cells and are not involved in neuronal signaling. Here, we report the identification of HVRP1, a protein related to the Hv1 channel (from which the name Hv1 Related Protein 1 is derived), which we find to be expressed primarily in the central nervous system, and particularly in the cerebellum. Within the cerebellar tissue, HVRP1 is specifically expressed in granule neurons, as determined by in situ hybridization and immunohistochemistry. Analysis of subcellular distribution via electron microscopy and immunogold labeling reveals that the protein localizes on the post-synaptic side of contacts between glutamatergic mossy fibers and the granule cells. We also find that, despite the similarities in amino acid sequence and structural organization between Hv1 and HVRP1, the two proteins have distinct functional properties. The high conservation of HVRP1 in vertebrates and its cellular and subcellular localizations suggest an important function in the nervous system.


Asunto(s)
Cerebelo/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Densidad Postsináptica/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Microscopía Electrónica , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Filogenia
20.
Proc Natl Acad Sci U S A ; 111(27): 9971-6, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24912149

RESUMEN

The voltage-gated proton channel Hv1 plays important roles in proton extrusion, pH homeostasis, and production of reactive oxygen species in a variety of cell types. Excessive Hv1 activity increases proliferation and invasiveness in cancer cells and worsens brain damage in ischemic stroke. The channel is composed of two subunits, each containing a proton-permeable voltage-sensing domain (VSD) and lacking the pore domain typical of other voltage-gated ion channels. We have previously shown that the compound 2-guanidinobenzimidazole (2GBI) inhibits Hv1 proton conduction by binding to the VSD from its intracellular side. Here, we examine the binding affinities of a series of 2GBI derivatives on human Hv1 channels mutated at positions located in the core of the VSD and apply mutant cycle analysis to determine how the inhibitor interacts with the channel. We identify four Hv1 residues involved in the binding: aspartate 112, phenylalanine 150, serine 181, and arginine 211. 2GBI appears to be oriented in the binding site with its benzo ring pointing to F150, its imidazole ring inserted between residue D112 and residues S181 and R211, and the guanidine group positioned in the proximity of R211. We also identify a modified version of 2GBI that is able to reach the binding site on Hv1 from the extracellular side of the membrane. Understanding how compounds like 2GBI interact with the Hv1 channel is an important step to the development of pharmacological treatments for diseases caused by Hv1 hyperactivity.


Asunto(s)
Guanidinas/farmacología , Canales Iónicos/antagonistas & inhibidores , Animales , Células Cultivadas , Humanos , Activación del Canal Iónico , Canales Iónicos/química , Canales Iónicos/fisiología , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...