Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(5): 1101-12, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26317472

RESUMEN

Potassium is the most abundant ion to face both plasma and organelle membranes. Extensive research over the past seven decades has characterized how K(+) permeates the plasma membrane to control fundamental processes such as secretion, neuronal communication, and heartbeat. However, how K(+) permeates organelles such as lysosomes and endosomes is unknown. Here, we directly recorded organelle K(+) conductance and discovered a major K(+)-selective channel KEL on endosomes and lysosomes. KEL is formed by TMEM175, a protein with unknown function. Unlike any of the ∼80 plasma membrane K(+) channels, TMEM175 has two repeats of 6-transmembrane-spanning segments and has no GYG K(+) channel sequence signature-containing, pore-forming P loop. Lysosomes lacking TMEM175 exhibit no K(+) conductance, have a markedly depolarized ΔΨ and little sensitivity to changes in [K(+)], and have compromised luminal pH stability and abnormal fusion with autophagosomes during autophagy. Thus, TMEM175 comprises a K(+) channel that underlies the molecular mechanism of lysosomal K(+) permeability.


Asunto(s)
Endosomas/metabolismo , Lisosomas/metabolismo , Canales de Potasio/metabolismo , Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Membranas Intracelulares/metabolismo , Ratones , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Fagosomas/metabolismo , Canales de Potasio/química , Canales de Potasio/genética , Alineación de Secuencia
2.
Cell ; 162(4): 836-48, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26276633

RESUMEN

Circadian clocks regulate membrane excitability in master pacemaker neurons to control daily rhythms of sleep and wake. Here, we find that two distinctly timed electrical drives collaborate to impose rhythmicity on Drosophila clock neurons. In the morning, a voltage-independent sodium conductance via the NA/NALCN ion channel depolarizes these neurons. This current is driven by the rhythmic expression of NCA localization factor-1, linking the molecular clock to ion channel function. In the evening, basal potassium currents peak to silence clock neurons. Remarkably, daily antiphase cycles of sodium and potassium currents also drive mouse clock neuron rhythms. Thus, we reveal an evolutionarily ancient strategy for the neural mechanisms that govern daily sleep and wake.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Drosophila/fisiología , Animales , Relojes Biológicos , Membrana Celular/metabolismo , Drosophila/citología , Proteínas de Drosophila/metabolismo , Técnicas de Silenciamiento del Gen , Canales Iónicos/genética , Canales Iónicos/metabolismo , Proteínas de la Membrana , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Sodio/metabolismo
3.
Cell ; 152(4): 778-790, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23394946

RESUMEN

Survival in the wild requires organismal adaptations to the availability of nutrients. Endosomes and lysosomes are key intracellular organelles that couple nutrition and metabolic status to cellular responses, but how they detect cytosolic ATP levels is not well understood. Here, we identify an endolysosomal ATP-sensitive Na(+) channel (lysoNa(ATP)). The channel is a complex formed by two-pore channels (TPC1 and TPC2), ion channels previously thought to be gated by nicotinic acid adenine dinucleotide phosphate (NAADP), and the mammalian target of rapamycin (mTOR). The channel complex detects nutrient status, becomes constitutively open upon nutrient removal and mTOR translocation off the lysosomal membrane, and controls the lysosome's membrane potential, pH stability, and amino acid homeostasis. Mutant mice lacking lysoNa(ATP) have much reduced exercise endurance after fasting. Thus, TPCs make up an ion channel family that couples the cell's metabolic state to endolysosomal function and are crucial for physical endurance during food restriction.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canales de Calcio/metabolismo , Lisosomas/metabolismo , Canales de Sodio/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Adenilato Quinasa/metabolismo , Aminoácidos/metabolismo , Animales , Canales de Calcio/química , Canales de Calcio/genética , Ayuno , Técnicas de Inactivación de Genes , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Potenciales de la Membrana , Ratones , Resistencia Física
4.
Cell ; 151(2): 372-83, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23063126

RESUMEN

Mammalian two-pore channel proteins (TPC1, TPC2; TPCN1, TPCN2) encode ion channels in intracellular endosomes and lysosomes and were proposed to mediate endolysosomal calcium release triggered by the second messenger, nicotinic acid adenine dinucleotide phosphate (NAADP). By directly recording TPCs in endolysosomes from wild-type and TPC double-knockout mice, here we show that, in contrast to previous conclusions, TPCs are in fact sodium-selective channels activated by PI(3,5)P(2) and are not activated by NAADP. Moreover, the primary endolysosomal ion is Na(+), not K(+), as had been previously assumed. These findings suggest that the organellar membrane potential may undergo large regulatory changes and may explain the specificity of PI(3,5)P(2) in regulating the fusogenic potential of intracellular organelles.


Asunto(s)
Canales de Calcio/metabolismo , Lisosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/genética , Línea Celular , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , NADP/análogos & derivados , NADP/metabolismo , Canales de Sodio/metabolismo
5.
Nature ; 591(7850): 431-437, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33505021

RESUMEN

Lysosomes have fundamental physiological roles and have previously been implicated in Parkinson's disease1-5. However, how extracellular growth factors communicate with intracellular organelles to control lysosomal function is not well understood. Here we report a lysosomal K+ channel complex that is activated by growth factors and gated by protein kinase B (AKT) that we term lysoKGF. LysoKGF consists of a pore-forming protein TMEM175 and AKT: TMEM175 is opened by conformational changes in, but not the catalytic activity of, AKT. The minor allele at rs34311866, a common variant in TMEM175, is associated with an increased risk of developing Parkinson's disease and reduces channel currents. Reduction in lysoKGF function predisposes neurons to stress-induced damage and accelerates the accumulation of pathological α-synuclein. By contrast, the minor allele at rs3488217-another common variant of TMEM175, which is associated with a decreased risk of developing Parkinson's disease-produces a gain-of-function in lysoKGF during cell starvation, and enables neuronal resistance to damage. Deficiency in TMEM175 leads to a loss of dopaminergic neurons and impairment in motor function in mice, and a TMEM175 loss-of-function variant is nominally associated with accelerated rates of cognitive and motor decline in humans with Parkinson's disease. Together, our studies uncover a pathway by which extracellular growth factors regulate intracellular organelle function, and establish a targetable mechanism by which common variants of TMEM175 confer risk for Parkinson's disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Canales de Potasio/metabolismo , Potasio/metabolismo , Animales , Biocatálisis , Neuronas Dopaminérgicas/metabolismo , Femenino , Mutación con Ganancia de Función , Células HEK293 , Humanos , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Noqueados , Destreza Motora , Complejos Multiproteicos/química , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Enfermedad de Parkinson/genética , Canales de Potasio/química , Canales de Potasio/deficiencia , Canales de Potasio/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , alfa-Sinucleína/metabolismo
6.
Proc Natl Acad Sci U S A ; 121(8): e2309465121, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38354262

RESUMEN

Phagocytes promptly resolve ingested targets to replenish lysosomes and maintain their responsiveness. The resolution process requires that degradative hydrolases, solute transporters, and proteins involved in lipid traffic are delivered and made active in phagolysosomes. It also involves extensive membrane remodeling. We report that cation channels that localize to phagolysosomes were essential for resolution. Specifically, the conductance of Na+ by two-pore channels (TPCs) and the presence of a Na+ gradient between the phagolysosome lumen and the cytosol were critical for the controlled release of membrane tension that permits deformation of the limiting phagolysosome membrane. In turn, membrane deformation was a necessary step to efficiently transport the cholesterol extracted from cellular targets, permeabilizing them to hydrolases. These results place TPCs as regulators of endomembrane remodeling events that precede target degradation in cases when the target is bound by a cholesterol-containing membrane. The findings may help to explain lipid metabolism dysfunction and autophagic flux impairment reported in TPC KO mice and establish stepwise regulation to the resolution process that begins with lysis of the target.


Asunto(s)
Fagosomas , Canales de Dos Poros , Ratones , Animales , Fagosomas/metabolismo , Lisosomas/metabolismo , Hidrolasas/metabolismo , Colesterol/metabolismo
7.
Annu Rev Pharmacol Toxicol ; 63: 19-41, 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36151054

RESUMEN

Lysosomes play fundamental roles in material digestion, cellular clearance, recycling, exocytosis, wound repair, Ca2+ signaling, nutrient signaling, and gene expression regulation. The organelle also serves as a hub for important signaling networks involving the mTOR and AKT kinases. Electrophysiological recording and molecular and structural studies in the past decade have uncovered several unique lysosomal ion channels and transporters, including TPCs, TMEM175, TRPMLs, CLN7, and CLC-7. They underlie the organelle's permeability to major ions, including K+, Na+, H+, Ca2+, and Cl-. The channels are regulated by numerous cellular factors, ranging from H+ in the lumen and voltage across the lysosomal membrane to ATP in the cytosol to growth factors outside the cell. Genetic variations in the channel/transporter genes are associated with diseases that include lysosomal storage diseases and neurodegenerative diseases. Recent studies with human genetics and channel activators suggest that lysosomal channels may be attractive targets for the development of therapeutics for the prevention of and intervention in human diseases.


Asunto(s)
Canales Iónicos , Enfermedades Neurodegenerativas , Humanos , Canales Iónicos/metabolismo , Transducción de Señal , Lisosomas/química , Lisosomas/metabolismo , Enfermedades Neurodegenerativas/metabolismo
8.
Genet Med ; 25(9): 100894, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37183800

RESUMEN

PURPOSE: The "NALCN channelosome" is an ion channel complex that consists of multiple proteins, including NALCN, UNC79, UNC80, and FAM155A. Only a small number of individuals with a neurodevelopmental syndrome have been reported with disease causing variants in NALCN and UNC80. However, no pathogenic UNC79 variants have been reported, and in vivo function of UNC79 in humans is largely unknown. METHODS: We used international gene-matching efforts to identify patients harboring ultrarare heterozygous loss-of-function UNC79 variants and no other putative responsible genes. We used genetic manipulations in Drosophila and mice to test potential causal relationships between UNC79 variants and the pathology. RESULTS: We found 6 unrelated and affected patients with UNC79 variants. Five patients presented with overlapping neurodevelopmental features, including mild to moderate intellectual disability and a mild developmental delay, whereas a single patient reportedly had normal cognitive and motor development but was diagnosed with epilepsy and autistic features. All displayed behavioral issues and 4 patients had epilepsy. Drosophila with UNC79 knocked down displayed induced seizure-like phenotype. Mice with a heterozygous loss-of-function variant have a developmental delay in body weight compared with wild type. In addition, they have impaired ability in learning and memory. CONCLUSION: Our results demonstrate that heterozygous loss-of-function UNC79 variants are associated with neurologic pathologies.


Asunto(s)
Epilepsia , Discapacidad Intelectual , Proteínas de la Membrana , Trastornos del Neurodesarrollo , Animales , Humanos , Ratones , Drosophila/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Trastornos del Neurodesarrollo/genética , Fenotipo , Proteínas de la Membrana/genética
9.
Nature ; 547(7664): 472-475, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28723891

RESUMEN

TMEM175 is a lysosomal K+ channel that is important for maintaining the membrane potential and pH stability in lysosomes. It contains two homologous copies of a six-transmembrane-helix (6-TM) domain, which has no sequence homology to the canonical tetrameric K+ channels and lacks the TVGYG selectivity filter motif found in these channels. The prokaryotic TMEM175 channel, which is present in a subset of bacteria and archaea, contains only a single 6-TM domain and functions as a tetramer. Here, we present the crystal structure of a prokaryotic TMEM175 channel from Chamaesiphon minutus, CmTMEM175, the architecture of which represents a completely different fold from that of canonical K+ channels. All six transmembrane helices of CmTMEM175 are tightly packed within each subunit without undergoing domain swapping. The highly conserved TM1 helix acts as the pore-lining inner helix, creating an hourglass-shaped ion permeation pathway in the channel tetramer. Three layers of hydrophobic residues on the carboxy-terminal half of the TM1 helices form a bottleneck along the ion conduction pathway and serve as the selectivity filter of the channel. Mutagenesis analysis suggests that the first layer of the highly conserved isoleucine residues in the filter is primarily responsible for channel selectivity. Thus, the structure of CmTMEM175 represents a novel architecture of a tetrameric cation channel whose ion selectivity mechanism appears to be distinct from that of the classical K+ channel family.


Asunto(s)
Lisosomas/química , Canales de Potasio/química , Canales de Potasio/metabolismo , Estructura Cuaternaria de Proteína , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Isoleucina/metabolismo , Modelos Moleculares
10.
Nature ; 550(7676): 411-414, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29019979

RESUMEN

The modulation of ion channel activity by lipids is increasingly recognized as a fundamental component of cellular signalling. The transient receptor potential mucolipin (TRPML) channel family belongs to the TRP superfamily and is composed of three members: TRPML1-TRPML3. TRPMLs are the major Ca2+-permeable channels on late endosomes and lysosomes (LEL). They regulate the release of Ca2+ from organelles, which is important for various physiological processes, including organelle trafficking and fusion. Loss-of-function mutations in the MCOLN1 gene, which encodes TRPML1, cause the neurodegenerative lysosomal storage disorder mucolipidosis type IV, and a gain-of-function mutation (Ala419Pro) in TRPML3 gives rise to the varitint-waddler (Va) mouse phenotype. Notably, TRPML channels are activated by the low-abundance and LEL-enriched signalling lipid phosphatidylinositol-3,5-bisphosphate (PtdIns(3,5)P2), whereas other phosphoinositides such as PtdIns(4,5)P2, which is enriched in plasma membranes, inhibit TRPMLs. Conserved basic residues at the N terminus of the channel are important for activation by PtdIns(3,5)P2 and inhibition by PtdIns(4,5)P2. However, owing to a lack of structural information, the mechanism by which TRPML channels recognize PtdIns(3,5)P2 and increase their Ca2+ conductance remains unclear. Here we present the cryo-electron microscopy (cryo-EM) structure of a full-length TRPML3 channel from the common marmoset (Callithrix jacchus) at an overall resolution of 2.9 Å. Our structure reveals not only the molecular basis of ion conduction but also the unique architecture of TRPMLs, wherein the voltage sensor-like domain is linked to the pore via a cytosolic domain that we term the mucolipin domain. Combined with functional studies, these data suggest that the mucolipin domain is responsible for PtdIns(3,5)P2 binding and subsequent channel activation, and that it acts as a 'gating pulley' for lipid-dependent TRPML gating.


Asunto(s)
Microscopía por Crioelectrón , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/ultraestructura , Animales , Sitios de Unión , Callithrix , Transporte Iónico , Modelos Moleculares , Fosfatos de Fosfatidilinositol/metabolismo , Dominios Proteicos , Canales de Potencial de Receptor Transitorio/metabolismo
11.
Nature ; 531(7593): 196-201, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26689363

RESUMEN

Two-pore channels (TPCs) contain two copies of a Shaker-like six-transmembrane (6-TM) domain in each subunit and are ubiquitously expressed in both animals and plants as organellar cation channels. Here we present the crystal structure of a vacuolar two-pore channel from Arabidopsis thaliana, AtTPC1, which functions as a homodimer. AtTPC1 activation requires both voltage and cytosolic Ca(2+). Ca(2+) binding to the cytosolic EF-hand domain triggers conformational changes coupled to the pair of pore-lining inner helices from the first 6-TM domains, whereas membrane potential only activates the second voltage-sensing domain, the conformational changes of which are coupled to the pair of inner helices from the second 6-TM domains. Luminal Ca(2+) or Ba(2+) can modulate voltage activation by stabilizing the second voltage-sensing domain in the resting state and shift voltage activation towards more positive potentials. Our Ba(2+)-bound AtTPC1 structure reveals a voltage sensor in the resting state, providing hitherto unseen structural insight into the general voltage-gating mechanism among voltage-gated channels.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/química , Canales de Calcio/química , Secuencia de Aminoácidos , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Bario/metabolismo , Sitios de Unión , Calcio/metabolismo , Calcio/farmacología , Canales de Calcio/genética , Canales de Calcio/metabolismo , Cristalografía por Rayos X , Citosol/metabolismo , Motivos EF Hand , Conductividad Eléctrica , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
13.
Am J Hum Genet ; 98(1): 202-9, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26708751

RESUMEN

Ion channel proteins are required for both the establishment of resting membrane potentials and the generation of action potentials. Hundreds of mutations in genes encoding voltage-gated ion channels responsible for action potential generation have been found to cause severe neurological diseases. In contrast, the roles of voltage-independent "leak" channels, important for the establishment and maintenance of resting membrane potentials upon which action potentials are generated, are not well established in human disease. UNC80 is a large component of the NALCN sodium-leak channel complex that regulates the basal excitability of the nervous system. Loss-of-function mutations of NALCN cause infantile hypotonia with psychomotor retardation and characteristic facies (IHPRF). We report four individuals from three unrelated families who have homozygous missense or compound heterozygous truncating mutations in UNC80 and persistent hypotonia, encephalopathy, growth failure, and severe intellectual disability. Compared to control cells, HEK293T cells transfected with an expression plasmid containing the c.5098C>T (p.Pro1700Ser) UNC80 mutation found in one individual showed markedly decreased NALCN channel currents. Our findings demonstrate the fundamental significance of UNC80 and basal ionic conductance to human health.


Asunto(s)
Alelos , Encefalopatías/genética , Proteínas Portadoras/genética , Trastornos del Crecimiento/genética , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Hipotonía Muscular/genética , Mutación , Adolescente , Niño , Preescolar , Femenino , Humanos , Índice de Severidad de la Enfermedad
14.
Annu Rev Physiol ; 77: 57-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25668017

RESUMEN

Lysosomes are acidic compartments filled with more than 60 different types of hydrolases. They mediate the degradation of extracellular particles from endocytosis and of intracellular components from autophagy. The digested products are transported out of the lysosome via specific catabolite exporters or via vesicular membrane trafficking. Lysosomes also contain more than 50 membrane proteins and are equipped with the machinery to sense nutrient availability, which determines the distribution, number, size, and activity of lysosomes to control the specificity of cargo flux and timing (the initiation and termination) of degradation. Defects in degradation, export, or trafficking result in lysosomal dysfunction and lysosomal storage diseases (LSDs). Lysosomal channels and transporters mediate ion flux across perimeter membranes to regulate lysosomal ion homeostasis, membrane potential, catabolite export, membrane trafficking, and nutrient sensing. Dysregulation of lysosomal channels underlies the pathogenesis of many LSDs and possibly that of metabolic and common neurodegenerative diseases.


Asunto(s)
Exocitosis/fisiología , Canales Iónicos/fisiología , Lisosomas/fisiología , Animales , Homeostasis/fisiología , Humanos , Iones/metabolismo , Enfermedades por Almacenamiento Lisosomal/fisiopatología , Potenciales de la Membrana/fisiología
15.
Nat Chem Biol ; 10(6): 463-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24776928

RESUMEN

The physiological function and molecular regulation of plasma membrane potential have been extensively studied, but how intracellular organelles sense and control membrane potential is not well understood. Using whole-organelle patch clamp recording, we show that endosomes and lysosomes are electrically excitable organelles. In a subpopulation of endolysosomes, a brief electrical stimulus elicits a prolonged membrane potential depolarization spike. The organelles have a previously uncharacterized, depolarization-activated, noninactivating Na(+) channel (lysoNaV). The channel is formed by a two-repeat six-transmembrane-spanning (2×6TM) protein, TPC1, which represents the evolutionary transition between 6TM and 4×6TM voltage-gated channels. Luminal alkalization also opens lysoNaV by markedly shifting the channel's voltage dependence of activation toward hyperpolarization. Thus, TPC1 is a member of a new family of voltage-gated Na(+) channels that senses pH changes and confers electrical excitability to organelles.


Asunto(s)
Canales de Calcio/fisiología , Endosomas/metabolismo , Activación del Canal Iónico/fisiología , Lisosomas/metabolismo , Canales de Sodio Activados por Voltaje/fisiología , Secuencia de Aminoácidos , Animales , Canales de Calcio/genética , Estimulación Eléctrica , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Membranas Intracelulares/metabolismo , Macrófagos Peritoneales/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Permeabilidad , Alineación de Secuencia , Canales de Sodio Activados por Voltaje/genética
16.
Annu Rev Physiol ; 74: 453-75, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22017176

RESUMEN

Ion channels control the sperm ability to fertilize the egg by regulating sperm maturation in the female reproductive tract and by triggering key sperm physiological responses required for successful fertilization such as hyperactivated motility, chemotaxis, and the acrosome reaction. CatSper, a pH-regulated, calcium-selective ion channel, and KSper (Slo3) are core regulators of sperm tail calcium entry and sperm hyperactivated motility. Many other channels had been proposed as regulating sperm activity without direct measurements. With the development of the sperm patch-clamp technique, CatSper and KSper have been confirmed as the primary spermatozoan ion channels. In addition, the voltage-gated proton channel Hv1 has been identified in human sperm tail, and the P2X2 ion channel has been identified in the midpiece of mouse sperm. Mutations and deletions in sperm-specific ion channels affect male fertility in both mice and humans without affecting other physiological functions. The uniqueness of sperm ion channels makes them ideal pharmaceutical targets for contraception. In this review we discuss how ion channels regulate sperm physiology.


Asunto(s)
Fertilidad/fisiología , Canales Iónicos/fisiología , Espermatozoides/fisiología , Animales , Canales de Calcio/fisiología , Quimiotaxis/fisiología , Femenino , Humanos , Masculino , Ratones , Técnicas de Placa-Clamp , Capacitación Espermática/fisiología , Motilidad Espermática/fisiología , Espermatozoides/metabolismo , Espermatozoides/ultraestructura
17.
Nature ; 457(7230): 741-4, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19092807

RESUMEN

Several neurotransmitters act through G-protein-coupled receptors to evoke a 'slow' excitation of neurons. These include peptides, such as substance P and neurotensin, as well as acetylcholine and noradrenaline. Unlike the fast (approximately millisecond) ionotropic actions of small-molecule neurotransmitters, the slow excitation is not well understood at the molecular level, but can be mainly attributed to suppressing K(+) currents and/or activating a non-selective cation channel. The molecular identity of this cation channel has yet to be determined; similarly, how the channel is activated and its relative contribution to neuronal excitability induced by the neuropeptides are unknown. Here we show that, in the mouse hippocampal and ventral tegmental area neurons, substance P and neurotensin activate a channel complex containing NALCN and a large previously unknown protein UNC-80. The activation by substance P through TACR1 (a G-protein-coupled receptor for substance P) occurs by means of a unique mechanism: it does not require G-protein activation but is dependent on Src family kinases. These findings identify NALCN as the cation channel activated by substance P receptor, and suggest that UNC-80 and Src family kinases, rather than a G protein, are involved in the coupling from receptor to channel.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/farmacología , Animales , Línea Celular , Conductividad Eléctrica , Guanosina Trifosfato/metabolismo , Proteínas de Unión al GTP Heterotriméricas , Hipocampo/citología , Humanos , Canales Iónicos/agonistas , Canales Iónicos/genética , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neurotensina/farmacología , Receptores de Neuroquinina-1/metabolismo , Sustancia P/farmacología , Transfección , Área Tegmental Ventral/citología , Familia-src Quinasas/metabolismo
18.
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
19.
Cell Physiol Biochem ; 29(3-4): 501-10, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22508057

RESUMEN

Interstitial cells of Cajal (ICCs) are the pacemaking cells in the gastrointestinal muscles that generate the rhythmic oscillations in membrane potential known as slow waves. ICCs also mediate or transduce inputs from the enteric nervous system. Substance P (SubP) is a member of the family of mammalian tachykinin peptides that are predominantly released by enteric neurons. This study assessed the relationship of Na(+)-leak channel (NALCN) in the SubP-induced depolarization in pacemaking activity in the gastrointestinal tract. The patch-clamp technique for whole-cell recording was used in cultured cluster and single ICCs. Electrophysiological and pharmacological properties of SubP in ICC pacemaking activity were similar to those of NALCN. Reverse-transcription polymerase chain reaction, Western blotting, and immunohistochemistry all showed abundant and localized expression of NALCN messenger RNA and protein in mouse small intestine. NALCN is involved in the SubP-induced depolarization of intestinal pacemaking activity. The protein is a potential target for pharmacological treatment of motor disorders of the gut.


Asunto(s)
Células Intersticiales de Cajal/fisiología , Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Sodio/metabolismo , Sustancia P/farmacología , Animales , Relojes Biológicos , Western Blotting , Células Cultivadas , Fenómenos Electrofisiológicos , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/fisiología , Motilidad Gastrointestinal , Inmunohistoquímica , Células Intersticiales de Cajal/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/fisiología , Potenciales de la Membrana , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neurotransmisores/farmacología , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Sodio/fisiología
20.
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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA