Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(5)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36901904

RESUMEN

TRPV5 and TRPV6 are calcium-selective ion channels expressed at the apical membrane of epithelial cells. Important for systemic calcium (Ca2+) homeostasis, these channels are considered gatekeepers of this cation transcellular transport. Intracellular Ca2+ exerts a negative control over the activity of these channels by promoting inactivation. TRPV5 and TRPV6 inactivation has been divided into fast and slow phases based on their kinetics. While slow inactivation is common to both channels, fast inactivation is characteristic of TRPV6. It has been proposed that the fast phase depends on Ca2+ binding and that the slow phase depends on the binding of the Ca2+/Calmodulin complex to the internal gate of the channels. Here, by means of structural analyses, site-directed mutagenesis, electrophysiology, and molecular dynamic simulations, we identified a specific set of amino acids and interactions that determine the inactivation kinetics of mammalian TRPV5 and TRPV6 channels. We propose that the association between the intracellular helix-loop-helix (HLH) domain and the TRP domain helix (TDh) favors the faster inactivation kinetics observed in mammalian TRPV6 channels.


Asunto(s)
Calcio , Canales Catiónicos TRPV , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Células Epiteliales/metabolismo , Secuencias Hélice-Asa-Hélice , Mamíferos/metabolismo , Canales Catiónicos TRPV/metabolismo , Humanos
2.
Elife ; 122023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36695574

RESUMEN

The severe acute respiratory syndrome associated coronavirus 2 (SARS-CoV-2) and SARS-CoV-1 accessory protein Orf3a colocalizes with markers of the plasma membrane, endocytic pathway, and Golgi apparatus. Some reports have led to annotation of both Orf3a proteins as viroporins. Here, we show that neither SARS-CoV-2 nor SARS-CoV-1 Orf3a form functional ion conducting pores and that the conductances measured are common contaminants in overexpression and with high levels of protein in reconstitution studies. Cryo-EM structures of both SARS-CoV-2 and SARS-CoV-1 Orf3a display a narrow constriction and the presence of a positively charged aqueous vestibule, which would not favor cation permeation. We observe enrichment of the late endosomal marker Rab7 upon SARS-CoV-2 Orf3a overexpression, and co-immunoprecipitation with VPS39. Interestingly, SARS-CoV-1 Orf3a does not cause the same cellular phenotype as SARS-CoV-2 Orf3a and does not interact with VPS39. To explain this difference, we find that a divergent, unstructured loop of SARS-CoV-2 Orf3a facilitates its binding with VPS39, a HOPS complex tethering protein involved in late endosome and autophagosome fusion with lysosomes. We suggest that the added loop enhances SARS-CoV-2 Orf3a's ability to co-opt host cellular trafficking mechanisms for viral exit or host immune evasion.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/metabolismo , Endosomas/metabolismo , Canales Iónicos/metabolismo
3.
Elife ; 112022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35686986

RESUMEN

Transient receptor potential (TRP) proteins are a large family of cation-selective channels, surpassed in variety only by voltage-gated potassium channels. Detailed molecular mechanisms governing how membrane voltage, ligand binding, or temperature can induce conformational changes promoting the open state in TRP channels are still a matter of debate. Aiming to unveil distinctive structural features common to the transmembrane domains within the TRP family, we performed phylogenetic reconstruction, sequence statistics, and structural analysis over a large set of TRP channel genes. Here, we report an exceptionally conserved set of residues. This fingerprint is composed of twelve residues localized at equivalent three-dimensional positions in TRP channels from the different subtypes. Moreover, these amino acids are arranged in three groups, connected by a set of aromatics located at the core of the transmembrane structure. We hypothesize that differences in the connectivity between these different groups of residues harbor the apparent differences in coupling strategies used by TRP subgroups.


Asunto(s)
Canales de Potencial de Receptor Transitorio , Filogenia , Dominios Proteicos , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/genética
4.
bioRxiv ; 2022 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-36263072

RESUMEN

The severe acute respiratory syndrome associated coronavirus 2 (SARS-CoV-2) and SARS-CoV-1 accessory protein Orf3a colocalizes with markers of the plasma membrane, endocytic pathway, and Golgi apparatus. Some reports have led to annotation of both Orf3a proteins as a viroporin. Here we show that neither SARS-CoV-2 nor SARS-CoV-1 form functional ion conducting pores and that the conductances measured are common contaminants in overexpression and with high levels of protein in reconstitution studies. Cryo-EM structures of both SARS-CoV-2 and SARS-CoV-1 Orf3a display a narrow constriction and the presence of a basic aqueous vestibule, which would not favor cation permeation. We observe enrichment of the late endosomal marker Rab7 upon SARS-CoV-2 Orf3a overexpression, and co-immunoprecipitation with VPS39. Interestingly, SARS-CoV-1 Orf3a does not cause the same cellular phenotype as SARS-CoV-2 Orf3a and does not interact with VPS39. To explain this difference, we find that a divergent, unstructured loop of SARS-CoV-2 Orf3a facilitates its binding with VPS39, a HOPS complex tethering protein involved in late endosome and autophagosome fusion with lysosomes. We suggest that the added loop enhances SARS-CoV-2 Orf3a ability to co-opt host cellular trafficking mechanisms for viral exit or host immune evasion.

5.
Elife ; 62017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29206105

RESUMEN

TRPV1 channels support the detection of noxious and nociceptive input. Currently available functional and structural data suggest that TRPV1 channels have two gates within their permeation pathway: one formed by a 'bundle-crossing' at the intracellular entrance and a second constriction at the selectivity filter. To describe conformational changes associated with channel gating, the fluorescent non-canonical amino acid coumarin-tyrosine was genetically encoded at Y671, a residue proximal to the selectivity filter. Total internal reflection fluorescence microscopy was performed to image the conformational dynamics of the channels in live cells. Photon counts and optical fluctuations from coumarin encoded within TRPV1 tetramers correlates with channel activation by capsaicin, providing an optical marker of conformational dynamics at the selectivity filter. In agreement with the fluorescence data, molecular dynamics simulations display alternating solvent exposure of Y671 in the closed and open states. Overall, the data point to a dynamic selectivity filter that may serve as a gate for permeation.


Cells use proteins on their surface as sensors to help them to assess and explore their environments and adapt to external conditions. The transient receptor potential (TRP) ion channels form one such family of proteins. Sodium, potassium and calcium ions can move through TRP channels to enter and exit cells, and by doing so trigger changes in the cell that help it respond to an external stimulus. The channels have physical "gates" that can open and close to control the flow of the ions. When the TRP channel detects a stimulus ­ which could take the form of specific chemicals, or a change in temperature, pressure or voltage ­ it changes shape, causing the gate to open. Researchers have a number of unanswered questions about how TRP channels work. Where in the channels are gates located? How do the channels control the flow of ions, and how do they communicate with each other? And which regions of the protein sense environmental cues? As a result, new technologies are being developed to make it easier to study the types of rearrangements that TRP channels experience when they activate. Steinberg, Kasimova et al. have used total internal reflection microscopy ­ a method that images fluorescent molecules ­ to measure the conformational change of a single TRP channel in a living cell. This channel, called TRPV1, senses external heat and plays an important role in pain perception. Its gate can also be opened by the pungent compound of chili pepper, capsaicin. The results of the experiments suggest that a selectivity filter region in TRPV1 channels changes its shape when the channel opens in response to capsaicin. Then, this selectivity filter appears to do double duty ­ it controls which types of ions pass through the channels as well as controlling their flow rate. Because of its role in pain perception, having a better understanding of how TRPV1 works will be valuable for researchers who are trying to develop new pain relief treatments. The so-called 'seeing is believing' method used by Steinberg, Kasimova et al. could also be used to study other membrane proteins, both to guide drug development and to improve our understanding of how cells interact with their environment.


Asunto(s)
Cumarinas/análisis , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/metabolismo , Tirosina/análisis , Capsaicina/metabolismo , Células HEK293 , Humanos , Microscopía Intravital , Microscopía Fluorescente , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación Proteica , Coloración y Etiquetado , Canales Catiónicos TRPV/genética
6.
Cell Rep ; 21(6): 1681-1691, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29117570

RESUMEN

Oxytocin is a hormone with various actions. Oxytocin-containing parvocellular neurons project to the brainstem and spinal cord. Oxytocin release from these neurons suppresses nociception of inflammatory pain, the molecular mechanism of which remains unclear. Here, we report that the noxious stimulus receptor TRPV1 is an ionotropic oxytocin receptor. Oxytocin elicits TRPV1 activity in native and heterologous expression systems, regardless of the presence of the classical oxytocin receptor. In TRPV1 knockout mice, DRG neurons exhibit reduced oxytocin sensitivity relative to controls, and oxytocin injections significantly attenuate capsaicin-induced nociception in in vivo experiments. Furthermore, oxytocin potentiates TRPV1 in planar lipid bilayers, supporting a direct agonistic action. Molecular modeling and simulation experiments provide insight into oxytocin-TRPV1 interactions, which resemble DkTx. Together, our findings suggest the existence of endogenous regulatory pathways that modulate nociception via direct action of oxytocin on TRPV1, implying its analgesic effect via channel desensitization.


Asunto(s)
Nocicepción/efectos de los fármacos , Oxitocina/farmacología , Canales Catiónicos TRPV/genética , Animales , Calcio/metabolismo , Capsaicina/análogos & derivados , Capsaicina/farmacología , Células Cultivadas , Potenciales Evocados/efectos de los fármacos , Femenino , Ganglios Espinales/citología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estructura Cuaternaria de Proteína , Receptores de Oxitocina/antagonistas & inhibidores , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo
7.
Curr Med Chem ; 22(25): 2943-53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26179995

RESUMEN

Melastatin-related Transient Receptor Potential 6 and 7 (TRPM6 and TRPM7) are cation channels with the almost unique trait of each possessing a kinase domain in its C terminus. Both the transmembrane pore and kinase are functional, and have been characterized experimentally, but whether one domain regulates the function of the other, or vice versa has remained largely unsettled. These proteins play important physiological roles in magnesium homeostasis and other cellular processes such as cell death, proliferation, differentiation and migration, and are consequently associated with several types of pathologies. Recently, studies performed in mice expressing a TRPM7 kinase-dead mutant suggest that the enzyme may function as part of a Mg(2+) sensor and transducer of signaling pathways during stressful environmental conditions. Additionally, it has been shown that TRPM7's kinase can act on its own in chromatin remodeling processes. Thus, the recent work in this field has provided new insights into the function of these interesting proteins and how they might be involved in human disease.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Humanos , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Canales Catiónicos TRPM/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA