Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.885
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(20): 4325-4344.e26, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37652010

RESUMEN

KCR channelrhodopsins (K+-selective light-gated ion channels) have received attention as potential inhibitory optogenetic tools but more broadly pose a fundamental mystery regarding how their K+ selectivity is achieved. Here, we present 2.5-2.7 Å cryo-electron microscopy structures of HcKCR1 and HcKCR2 and of a structure-guided mutant with enhanced K+ selectivity. Structural, electrophysiological, computational, spectroscopic, and biochemical analyses reveal a distinctive mechanism for K+ selectivity; rather than forming the symmetrical filter of canonical K+ channels achieving both selectivity and dehydration, instead, three extracellular-vestibule residues within each monomer form a flexible asymmetric selectivity gate, while a distinct dehydration pathway extends intracellularly. Structural comparisons reveal a retinal-binding pocket that induces retinal rotation (accounting for HcKCR1/HcKCR2 spectral differences), and design of corresponding KCR variants with increased K+ selectivity (KALI-1/KALI-2) provides key advantages for optogenetic inhibition in vitro and in vivo. Thus, discovery of a mechanism for ion-channel K+ selectivity also provides a framework for next-generation optogenetics.


Asunto(s)
Channelrhodopsins , Rhinosporidium , Humanos , Channelrhodopsins/química , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Channelrhodopsins/ultraestructura , Microscopía por Crioelectrón , Canales Iónicos , Potasio/metabolismo , Rhinosporidium/química
2.
Annu Rev Immunol ; 33: 291-353, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25861976

RESUMEN

Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata/fisiología , Canales Iónicos/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Humanos , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/metabolismo , Inmunoterapia/métodos , Canales Iónicos/genética , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Terapia Molecular Dirigida , Mutación , Transducción de Señal
3.
Cell ; 184(2): 534-544.e11, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33373586

RESUMEN

Determination of what is the specificity of subunits composing a protein complex is essential when studying gene variants on human pathophysiology. The pore-forming α-subunit KCNQ1, which belongs to the voltage-gated ion channel superfamily, associates to its ß-auxiliary subunit KCNE1 to generate the slow cardiac potassium IKs current, whose dysfunction leads to cardiac arrhythmia. Using pharmacology, gene invalidation, and single-molecule fluorescence assays, we found that KCNE1 fulfils all criteria of a bona fide auxiliary subunit of the TMEM16A chloride channel, which belongs to the anoctamin superfamily. Strikingly, assembly with KCNE1 switches TMEM16A from a calcium-dependent to a voltage-dependent ion channel. Importantly, clinically relevant inherited mutations within the TMEM16A-regulating domain of KCNE1 abolish the TMEM16A modulation, suggesting that the TMEM16A-KCNE1 current may contribute to inherited pathologies. Altogether, these findings challenge the dogma of the specificity of auxiliary subunits regarding protein complexes and questions ion channel classification.


Asunto(s)
Canales de Potasio con Entrada de Voltaje/metabolismo , Subunidades de Proteína/metabolismo , Animales , Anoctamina-1/metabolismo , Calcio/metabolismo , Canales de Cloruro/metabolismo , Células HEK293 , Humanos , Túbulos Renales Proximales/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Péptidos/metabolismo , Polimorfismo Genético , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Unión Proteica , Dominios Proteicos , Sistema Renina-Angiotensina
4.
Cell ; 173(4): 894-905.e13, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29706545

RESUMEN

Perceptual decisions require the accumulation of sensory information to a response criterion. Most accounts of how the brain performs this process of temporal integration have focused on evolving patterns of spiking activity. We report that subthreshold changes in membrane voltage can represent accumulating evidence before a choice. αß core Kenyon cells (αßc KCs) in the mushroom bodies of fruit flies integrate odor-evoked synaptic inputs to action potential threshold at timescales matching the speed of olfactory discrimination. The forkhead box P transcription factor (FoxP) sets neuronal integration and behavioral decision times by controlling the abundance of the voltage-gated potassium channel Shal (KV4) in αßc KC dendrites. αßc KCs thus tailor, through a particular constellation of biophysical properties, the generic process of synaptic integration to the demands of sequential sampling.


Asunto(s)
Dendritas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Bario/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Ciclohexanoles/farmacología , Proteínas de Drosophila/genética , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Masculino , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Receptores Odorantes/metabolismo , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Olfato , Sinapsis/metabolismo
5.
Immunity ; 55(8): 1466-1482.e9, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35863346

RESUMEN

Although many studies have addressed the regulatory circuits affecting neuronal activities, local non-synaptic mechanisms that determine neuronal excitability remain unclear. Here, we found that microglia prevented overactivation of pre-sympathetic neurons in the hypothalamic paraventricular nucleus (PVN) at steady state. Microglia constitutively released platelet-derived growth factor (PDGF) B, which signaled via PDGFRα on neuronal cells and promoted their expression of Kv4.3, a key subunit that conducts potassium currents. Ablation of microglia, conditional deletion of microglial PDGFB, or suppression of neuronal PDGFRα expression in the PVN elevated the excitability of pre-sympathetic neurons and sympathetic outflow, resulting in a profound autonomic dysfunction. Disruption of the PDGFBMG-Kv4.3Neuron pathway predisposed mice to develop hypertension, whereas central supplementation of exogenous PDGFB suppressed pressor response when mice were under hypertensive insult. Our results point to a non-immune action of resident microglia in maintaining the balance of sympathetic outflow, which is important in preventing cardiovascular diseases.


Asunto(s)
Hipertensión , Microglía , Animales , Hipertensión/metabolismo , Ratones , Neuronas/fisiología , Potasio/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
6.
Mol Cell ; 82(13): 2427-2442.e4, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35597238

RESUMEN

The voltage-gated ion channel activity depends on both activation (transition from the resting state to the open state) and inactivation. Inactivation is a self-restraint mechanism to limit ion conduction and is as crucial to membrane excitability as activation. Inactivation can occur when the channel is open or closed. Although open-state inactivation is well understood, the molecular basis of closed-state inactivation has remained elusive. We report cryo-EM structures of human KV4.2 channel complexes in inactivated, open, and closed states. Closed-state inactivation of KV4 involves an unprecedented symmetry breakdown for pore closure by only two of the four S4-S5 linkers, distinct from known mechanisms of open-state inactivation. We further capture KV4 in a putative resting state, revealing how voltage sensor movements control the pore. Moreover, our structures provide insights regarding channel modulation by KChIP2 and DPP6 auxiliary subunits. Our findings elucidate mechanisms of closed-state inactivation and voltage-dependent activation of the KV4 channel.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio Shal , Humanos , Activación del Canal Iónico/fisiología , Cinética , Potenciales de la Membrana/fisiología , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo
7.
Annu Rev Neurosci ; 44: 335-357, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-33770451

RESUMEN

The large number of ion channels found in all nervous systems poses fundamental questions concerning how the characteristic intrinsic properties of single neurons are determined by the specific subsets of channels they express. All neurons display many different ion channels with overlapping voltage- and time-dependent properties. We speculate that these overlapping properties promote resilience in neuronal function. Individual neurons of the same cell type show variability in ion channel conductance densities even though they can generate reliable and similar behavior. This complicates a simple assignment of function to any conductance and is associated with variable responses of neurons of the same cell type to perturbations, deletions, and pharmacological manipulation. Ion channel genes often show strong positively correlated expression, which may result from the molecular and developmental rules that determine which ion channels are expressed in a given cell type.


Asunto(s)
Canales Iónicos , Neuronas
8.
Annu Rev Cell Dev Biol ; 31: 231-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26566112

RESUMEN

Ion channels have emerged as regulators of developmental processes. In model organisms and in people with mutations in ion channels, disruption of ion channel function can affect cell proliferation, cell migration, and craniofacial and limb patterning. Alterations of ion channel function affect morphogenesis in fish, frogs, mammals, and flies, demonstrating that ion channels have conserved roles in developmental processes. One model suggests that ion channels affect proliferation and migration through changes in cell volume. However, ion channels have not explicitly been placed in canonical developmental signaling cascades until recently. This review gives examples of ion channels that influence developmental processes, offers a potential underlying molecular mechanism involving bone morphogenetic protein (BMP) signaling, and finally explores exciting possibilities for manipulating ion channels to influence cell fate for regenerative medicine and to impact disease.


Asunto(s)
Canales Iónicos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Tamaño de la Célula , Humanos , Transducción de Señal/fisiología
9.
Annu Rev Physiol ; 86: 277-300, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-37906945

RESUMEN

Novel KCNMA1 variants, encoding the BK K+ channel, are associated with a debilitating dyskinesia and epilepsy syndrome. Neurodevelopmental delay, cognitive disability, and brain and structural malformations are also diagnosed at lower incidence. More than half of affected individuals present with a rare negative episodic motor disorder, paroxysmal nonkinesigenic dyskinesia (PNKD3). The mechanistic relationship of PNKD3 to epilepsy and the broader spectrum of KCNMA1-associated symptomology is unknown. This review summarizes patient-associated KCNMA1 variants within the BK channel structure, functional classifications, genotype-phenotype associations, disease models, and treatment. Patient and transgenic animal data suggest delineation of gain-of-function (GOF) and loss-of-function KCNMA1 neurogenetic disease, validating two heterozygous alleles encoding GOF BK channels (D434G and N999S) as causing seizure and PNKD3. This discovery led to a variant-defined therapeutic approach for PNKD3, providing initial insight into the neurological basis. A comprehensive clinical definition of monogenic KCNMA1-linked disease and the neuronal mechanisms currently remain priorities for continued investigation.


Asunto(s)
Canalopatías , Corea , Epilepsia , Animales , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio , Canalopatías/genética , Epilepsia/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética
10.
Trends Biochem Sci ; 48(4): 331-344, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36336552

RESUMEN

The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome is a cytoplasmic supramolecular complex that is activated in response to cellular perturbations triggered by infection and sterile injury. Assembly of the NLRP3 inflammasome leads to activation of caspase-1, which induces the maturation and release of interleukin-1ß (IL-1ß) and IL-18, as well as cleavage of gasdermin D (GSDMD), which promotes a lytic form of cell death. Production of IL-1ß via NLRP3 can contribute to the pathogenesis of inflammatory disease, whereas aberrant IL-1ß secretion through inherited NLRP3 mutations causes autoinflammatory disorders. In this review, we discuss recent developments in the structure of the NLRP3 inflammasome, and the cellular processes and signaling events controlling its assembly and activation.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal , Caspasa 1/metabolismo , Expresión Génica , Interleucina-1beta/genética , Interleucina-1beta/metabolismo
11.
EMBO J ; 42(8): e112401, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-36811145

RESUMEN

The maintenance of sodium/potassium (Na+ /K+ ) homeostasis in plant cells is essential for salt tolerance. Plants export excess Na+ out of cells mainly through the Salt Overly Sensitive (SOS) pathway, activated by a calcium signal; however, it is unknown whether other signals regulate the SOS pathway and how K+ uptake is regulated under salt stress. Phosphatidic acid (PA) is emerging as a lipid signaling molecule that modulates cellular processes in development and the response to stimuli. Here, we show that PA binds to the residue Lys57 in SOS2, a core member of the SOS pathway, under salt stress, promoting the activity and plasma membrane localization of SOS2, which activates the Na+ /H+ antiporter SOS1 to promote the Na+ efflux. In addition, we reveal that PA promotes the phosphorylation of SOS3-like calcium-binding protein 8 (SCaBP8) by SOS2 under salt stress, which attenuates the SCaBP8-mediated inhibition of Arabidopsis K+ transporter 1 (AKT1), an inward-rectifying K+ channel. These findings suggest that PA regulates the SOS pathway and AKT1 activity under salt stress, promoting Na+ efflux and K+ influx to maintain Na+ /K+ homeostasis.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Proteínas Serina-Treonina Quinasas , Estrés Salino , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Homeostasis , Ácidos Fosfatidicos/metabolismo , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Salino/genética , Sodio/metabolismo
12.
Am J Hum Genet ; 111(4): 761-777, 2024 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-38503299

RESUMEN

Ion channels mediate voltage fluxes or action potentials that are central to the functioning of excitable cells such as neurons. The KCNB family of voltage-gated potassium channels (Kv) consists of two members (KCNB1 and KCNB2) encoded by KCNB1 and KCNB2, respectively. These channels are major contributors to delayed rectifier potassium currents arising from the neuronal soma which modulate overall excitability of neurons. In this study, we identified several mono-allelic pathogenic missense variants in KCNB2, in individuals with a neurodevelopmental syndrome with epilepsy and autism in some individuals. Recurrent dysmorphisms included a broad forehead, synophrys, and digital anomalies. Additionally, we selected three variants where genetic transmission has not been assessed, from two epilepsy studies, for inclusion in our experiments. We characterized channel properties of these variants by expressing them in oocytes of Xenopus laevis and conducting cut-open oocyte voltage clamp electrophysiology. Our datasets indicate no significant change in absolute conductance and conductance-voltage relationships of most disease variants as compared to wild type (WT), when expressed either alone or co-expressed with WT-KCNB2. However, variants c.1141A>G (p.Thr381Ala) and c.641C>T (p.Thr214Met) show complete abrogation of currents when expressed alone with the former exhibiting a left shift in activation midpoint when expressed alone or with WT-KCNB2. The variants we studied, nevertheless, show collective features of increased inactivation shifted to hyperpolarized potentials. We suggest that the effects of the variants on channel inactivation result in hyper-excitability of neurons, which contributes to disease manifestations.


Asunto(s)
Epilepsia , Mutación Missense , Trastornos del Neurodesarrollo , Canales de Potasio Shab , Animales , Humanos , Potenciales de Acción , Epilepsia/genética , Neuronas , Oocitos , Xenopus laevis , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Trastornos del Neurodesarrollo/genética
13.
Plant Cell ; 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38819305

RESUMEN

Potassium (K+) plays crucial roles in both plant development and immunity. However, the function of K+ in plant-virus interactions remains largely unknown. Here, we utilized Barley yellow striate mosaic virus (BYSMV), an insect-transmitted plant cytorhabdovirus, to investigate the interplay between viral infection and plant K+ homeostasis. The BYSMV accessory P9 protein exhibits viroporin activity by enhancing membrane permeability in Escherichia coli. Additionally, P9 increases K+ uptake in yeast (Saccharomyces cerevisiae) cells, which is disrupted by a point mutation of Glycine 14 to Threonine (P9G14T). Furthermore, BYSMV P9 forms oligomers and targets to both the viral envelope and the plant membrane. Based on the recombinant BYSMV-green fluorescent protein (BYGFP) virus, a P9-deleted mutant (BYGFPΔP9) was rescued and demonstrated infectivity within individual plant cells of Nicotiana benthamiana and insect vectors. However, BYGFPΔP9 failed to infect barley plants after transmission by insect vectors. Furthermore, infection of barley plants was severely impaired for BYGFP-P9G14T lacking P9 K+ channel activity. In vitro assays demonstrate that K+ facilitates virion disassembly and the release of genome RNA for viral mRNA transcription. Altogether, our results show that the K+ channel activity of viroporins is conserved in plant cytorhabdoviruses and plays crucial roles in insect-mediated virus transmission.

14.
Immunity ; 49(1): 56-65.e4, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29958799

RESUMEN

Potassium (K+) efflux across the plasma membrane is thought to be an essential mechanism for ATP-induced NLRP3 inflammasome activation, yet the identity of the efflux channel has remained elusive. Here we identified the two-pore domain K+ channel (K2P) TWIK2 as the K+ efflux channel triggering NLRP3 inflammasome activation. Deletion of Kcnk6 (encoding TWIK2) prevented NLRP3 activation in macrophages and suppressed sepsis-induced lung inflammation. Adoptive transfer of Kcnk6-/- macrophages into mouse airways after macrophage depletion also prevented inflammatory lung injury. The K+ efflux channel TWIK2 in macrophages has a fundamental role in activating the NLRP3 inflammasome and consequently mediates inflammation, pointing to TWIK2 as a potential target for anti-inflammatory therapies.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/fisiopatología , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Caspasa 1/deficiencia , Caspasa 1/metabolismo , Línea Celular , Inflamasomas/efectos de los fármacos , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/fisiopatología , Macrófagos/trasplante , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Quinina/farmacología , ARN Interferente Pequeño/farmacología , Receptores Purinérgicos P2X7/deficiencia , Receptores Purinérgicos P2X7/metabolismo , Sepsis/metabolismo , Sepsis/fisiopatología , Transducción de Señal/efectos de los fármacos
15.
Immunity ; 49(3): 413-426.e5, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30170814

RESUMEN

Inflammasome-activated caspase-1 cleaves gasdermin D to unmask its pore-forming activity, the predominant consequence of which is pyroptosis. Here, we report an additional biological role for gasdermin D in limiting cytosolic DNA surveillance. Cytosolic DNA is sensed by Aim2 and cyclic GMP-AMP synthase (cGAS) leading to inflammasome and type I interferon responses, respectively. We found that gasdermin D activated by the Aim2 inflammasome suppressed cGAS-driven type I interferon response to cytosolic DNA and Francisella novicida in macrophages. Similarly, interferon-ß (IFN-ß) response to F. novicida infection was elevated in gasdermin D-deficient mice. Gasdermin D-mediated negative regulation of IFN-ß occurred in a pyroptosis-, interleukin-1 (IL-1)-, and IL-18-independent manner. Mechanistically, gasdermin D depleted intracellular potassium (K+) via membrane pores, and this K+ efflux was necessary and sufficient to inhibit cGAS-dependent IFN-ß response. Thus, our findings have uncovered an additional interferon regulatory module involving gasdermin D and K+ efflux.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Francisella/fisiología , Infecciones por Bacterias Gramnegativas/inmunología , Inflamasomas/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Interferón Tipo I/metabolismo , Interleucina-1/metabolismo , Interleucina-18/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Proteínas de Unión a Fosfato , Potasio/metabolismo , ARN Interferente Pequeño/genética
16.
Proc Natl Acad Sci U S A ; 121(1): e2311402121, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38147555

RESUMEN

The planetesimals in the solar system exhibit varying degrees of moderately volatile elements (MVEs) depletion compared to the protosolar composition. Revealing the relevant mechanisms is crucial for exploring early solar system evolution. Most volatile-depleted materials in the solar system exhibit enrichments in the heavier isotopes of MVEs, which have traditionally been attributed to the loss of volatiles through partial evaporation. Angrites are so far an exception as they are enriched in the lighter isotopes of K. This has been interpreted as reflecting condensation processes. Here, we present Rb isotopic data of angrites and find that they have lighter Rb isotopic compositions than Vesta, Mars, and the Moon. The δ87Rb value of the angrite parent body (APB) is estimated to range between -1.19‰ and -0.67‰. The extremely light Rb isotopic composition of the APB is likely a result of the kinetic recondensation of Rb after near-complete evaporation during the magma ocean stage. This finding provides further support for the partial recondensation model to explain the light Rb and K isotopic compositions of the APB. In addition, the APB, alongside other terrestrial planetary bodies (e.g., Earth, Mars, Moon, and Vesta), exhibit a strong correlation between their Rb and K isotopic compositions. This coupling of Rb and K isotopes is indicative of a volatility-driven isotopic fractionation rather than nucleosynthetic anomalies. The extremely light Rb-K isotopic signatures of the APB suggest that beyond evaporation, condensation plays an equally significant role in shaping the planetary-scale distributions of volatile elements.

17.
Proc Natl Acad Sci U S A ; 121(30): e2407461121, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39018191

RESUMEN

The Shaker family of voltage-gated K+ channels has been thought of as an animal-specific ion channel family that diversified in concert with nervous systems. It comprises four functionally independent gene subfamilies (Kv1-4) that encode diverse neuronal K+ currents. Comparison of animal genomes predicts that only the Kv1 subfamily was present in the animal common ancestor. Here, we show that some choanoflagellates, the closest protozoan sister lineage to animals, also have Shaker family K+ channels. Choanoflagellate Shaker family channels are surprisingly most closely related to the animal Kv2-4 subfamilies which were believed to have evolved only after the divergence of ctenophores and sponges from cnidarians and bilaterians. Structural modeling predicts that the choanoflagellate channels share a T1 Zn2+ binding site with Kv2-4 channels that is absent in Kv1 channels. We functionally expressed three Shakers from Salpingoeca helianthica (SheliKvT1.1-3) in Xenopus oocytes. SheliKvT1.1-3 function only in two heteromultimeric combinations (SheliKvT1.1/1.2 and SheliKvT1.1/1.3) and encode fast N-type inactivating K+ channels with distinct voltage dependence that are most similar to the widespread animal Kv1-encoded A-type Shakers. Structural modeling of the T1 assembly domain supports a preference for heteromeric assembly in a 2:2 stoichiometry. These results push the origin of the Shaker family back into a common ancestor of metazoans and choanoflagellates. They also suggest that the animal common ancestor had at least two distinct molecular lineages of Shaker channels, a Kv1 subfamily lineage predicted from comparison of animal genomes and a Kv2-4 lineage predicted from comparison of animals and choanoflagellates.


Asunto(s)
Coanoflagelados , Evolución Molecular , Canales de Potasio de la Superfamilia Shaker , Animales , Coanoflagelados/genética , Coanoflagelados/metabolismo , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/metabolismo , Filogenia , Secuencia de Aminoácidos
18.
Proc Natl Acad Sci U S A ; 121(2): e2309579121, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38175865

RESUMEN

Nigericin, an ionophore derived from Streptomyces hygroscopicus, is arguably the most commonly used tool compound to study the NLRP3 inflammasome. Recent findings, however, showed that nigericin also activates the NLRP1 inflammasome in human keratinocytes. In this study, we resolve the mechanistic basis of nigericin-driven NLRP1 inflammasome activation. In multiple nonhematopoietic cell types, nigericin rapidly and specifically inhibits the elongation stage of the ribosome cycle by depleting cytosolic potassium ions. This activates the ribotoxic stress response (RSR) sensor kinase ZAKα, p38, and JNK, as well as the hyperphosphorylation of the NLRP1 linker domain. As a result, nigericin-induced pyroptosis in human keratinocytes is blocked by extracellular potassium supplementation, ZAKα knockout, or pharmacologic inhibitors of ZAKα and p38 kinase activities. By surveying a panel of ionophores, we show that electroneutrality of ion movement is essential to activate ZAKα-driven RSR and a greater extent of K+ depletion is necessary to activate ZAKα-NLRP1 than NLRP3. These findings resolve the mechanism by which nigericin activates NLRP1 in nonhematopoietic cell types and demonstrate an unexpected connection between RSR, perturbations of potassium ion flux, and innate immunity.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nigericina/farmacología , Potasio/metabolismo , Inmunidad Innata , Ionóforos , Proteínas NLR
19.
Proc Natl Acad Sci U S A ; 121(3): e2307776121, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38194456

RESUMEN

De novo heterozygous variants in KCNC2 encoding the voltage-gated potassium (K+) channel subunit Kv3.2 are a recently described cause of developmental and epileptic encephalopathy (DEE). A de novo variant in KCNC2 c.374G > A (p.Cys125Tyr) was identified via exome sequencing in a patient with DEE. Relative to wild-type Kv3.2, Kv3.2-p.Cys125Tyr induces K+ currents exhibiting a large hyperpolarizing shift in the voltage dependence of activation, accelerated activation, and delayed deactivation consistent with a relative stabilization of the open conformation, along with increased current density. Leveraging the cryogenic electron microscopy (cryo-EM) structure of Kv3.1, molecular dynamic simulations suggest that a strong π-π stacking interaction between the variant Tyr125 and Tyr156 in the α-6 helix of the T1 domain promotes a relative stabilization of the open conformation of the channel, which underlies the observed gain of function. A multicompartment computational model of a Kv3-expressing parvalbumin-positive cerebral cortex fast-spiking γ-aminobutyric acidergic (GABAergic) interneuron (PV-IN) demonstrates how the Kv3.2-Cys125Tyr variant impairs neuronal excitability and dysregulates inhibition in cerebral cortex circuits to explain the resulting epilepsy.


Asunto(s)
Epilepsia , Canales de Potasio Shaw , Humanos , Canales de Potasio Shaw/genética , Interneuronas , Corteza Cerebral , Epilepsia/genética , Mutación
20.
Proc Natl Acad Sci U S A ; 121(13): e2315407121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38502699

RESUMEN

Organic electrodes mainly consisting of C, O, H, and N are promising candidates for advanced batteries. However, the sluggish ionic and electronic conductivity limit the full play of their high theoretical capacities. Here, we integrate the idea of metal-support interaction in single-atom catalysts with π-d hybridization into the design of organic electrode materials for the applications of lithium (LIBs) and potassium-ion batteries (PIBs). Several types of transition metal single atoms (e.g., Co, Ni, Fe) with π-d hybridization are incorporated into the semiconducting covalent organic framework (COF) composite. Single atoms favorably modify the energy band structure and improve the electronic conductivity of COF. More importantly, the electronic interaction between single atoms and COF adjusts the binding affinity and modifies ion traffic between Li/K ions and the active organic units of COFs as evidenced by extensive in situ and ex situ characterizations and theoretical calculations. The corresponding LIB achieves a high reversible capacity of 1,023.0 mA h g-1 after 100 cycles at 100 mA g-1 and 501.1 mA h g-1 after 500 cycles at 1,000 mA g-1. The corresponding PIB delivers a high reversible capacity of 449.0 mA h g-1 at 100 mA g-1 after 150 cycles and stably cycled over 500 cycles at 1,000 mA g-1. This work provides a promising route to engineering organic electrodes.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA