Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38836042

RESUMEN

Objective: This study aimed to investigate the contamination status of hospital sinks with carbapenemase-producing Enterobacterales (CPE), the efficacy of daily cleaning with sodium hypochlorite, and the relationships between CPEs isolated from contaminated sinks and patients. Design: Pre/postintervention surveys of the CPE-contaminated sinks. Setting: Hospital wards including pediatric intensive care unit in a children's hospital. Participants: Consenting CPE-colonized patients admitted between November 2018 and June 2021 in our hospital. Methods: Environmental culture of 180 sinks from nine wards in our hospital was performed three times with an interval of 2 years (2019, 2021, 2023). Molecular typing of the isolated strains from the sinks and patients was performed. After the first surveillance culture, we initiated daily disinfection of the sinks using sodium hypochlorite. Results: Before the intervention, we detected 30 CPE-positive sinks in 2019. After the intervention with sodium hypochlorite, we observed a substantial decline in the number of sinks contaminated with CPE; 13 in 2021 and 6 in 2023. However, the intervention did not significantly reduce the number of CPE-contaminated sinks used for the disposal of nutrition-rich substances. The CPE isolates from the patients and those from the sinks of the wards or floors where they were admitted tended to have similar pulse-field gel electrophoresis patterns. Conclusion: Contaminated sinks could be reservoirs of disseminating CPE to the patients. Daily disinfection of sinks with sodium hypochlorite may be effective in eliminating CPE, although the effect could be weaker in sinks with a greater risk of contact with nutrition-rich substances.

2.
Environ Monit Assess ; 196(7): 616, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874785

RESUMEN

Forest pests pose a major threat to ecosystem services worldwide, requiring effective monitoring and management strategies. Recently, satellite remote sensing has emerged as a valuable tool to detect defoliation caused by these pests. Lymantria dispar, a major forest pest native to Japan, Siberia, and Europe, as well as introduced regions in North America, is of particular concern. In this study, we used Sentinel-2 satellite imagery to estimate the defoliation area and predict the distribution of L. dispar in Toyama Prefecture, central Japan. The primary aim was to understand the spatial distribution of L. dispar. The normalized difference vegetation index (NDVI) difference analysis estimated a defoliation area of 7.89 km2 in Toyama Prefecture for the year 2022. MaxEnt modeling, using defoliation map as occurrence data, identified the deciduous forests between approximately 35° and 50° at elevations of 400 m and 700 m as highly suitable for L. dispar. This predicted suitability was also high for larval locations but low for egg mass locations, likely due to differences in larval habitats and ovipositing sites. This study is the first attempt to utilize NDVI-based estimates as a proxy for MaxEnt. Our results showed higher prediction accuracy than a previous study based on the occurrence records including larvae, adults, and egg masses, indicating better discrimination of the distribution of L. dispar defoliation. Therefore, our approach to integrating satellite data and species distribution models can potentially enhance the assessment of areas affected by pests for effective forest management.


Asunto(s)
Monitoreo del Ambiente , Bosques , Animales , Monitoreo del Ambiente/métodos , Japón , Imágenes Satelitales , Tecnología de Sensores Remotos , Ecosistema , Mariposas Nocturnas/fisiología , Larva
3.
Elife ; 122023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36806330

RESUMEN

Ca2+ release-activated Ca2+ (CRAC) channels are activated by direct physical interactions between Orai1, the channel protein, and STIM1, the endoplasmic reticulum Ca2+ sensor. A hallmark of CRAC channels is fast Ca2+-dependent inactivation (CDI) which provides negative feedback to limit Ca2+ entry through CRAC channels. Although STIM1 is thought to be essential for CDI, its molecular mechanism remains largely unknown. Here, we examined a poorly understood gain-of-function (GOF) human Orai1 disease mutation, L138F, that causes tubular aggregate myopathy. Through pairwise mutational analysis, we determine that large amino acid substitutions at either L138 or the neighboring T92 locus located on the pore helix evoke highly Ca2+-selective currents in the absence of STIM1. We find that the GOF phenotype of the L138 pathogenic mutation arises due to steric clash between L138 and T92. Surprisingly, strongly activating L138 and T92 mutations showed CDI in the absence of STIM1, contradicting prevailing views that STIM1 is required for CDI. CDI of constitutively open T92W and L138F mutants showed enhanced intracellular Ca2+ sensitivity, which was normalized by re-adding STIM1 to the cells. Truncation of the Orai1 C-terminus reduced T92W CDI, indicating a key role for the Orai1 C-terminus for CDI. Overall, these results identify the molecular basis of a disease phenotype with broad implications for activation and inactivation of Orai1 channels.


Asunto(s)
Canales de Calcio , Canales de Calcio Activados por la Liberación de Calcio , Humanos , Canales de Calcio/metabolismo , Proteína ORAI1/genética , Mutación , Canales de Calcio Activados por la Liberación de Calcio/genética , Mutación con Ganancia de Función , Molécula de Interacción Estromal 1/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
4.
Sci Adv ; 9(4): eade7002, 2023 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-36706180

RESUMEN

Microglia are important mediators of neuroinflammation, which underlies neuropathic pain. However, the molecular checkpoints controlling microglial reactivity are not well-understood. Here, we investigated the role of Orai1 channels for microglia-mediated neuroinflammation following nerve injury and find that deletion of Orai1 in microglia attenuates Ca2+ signaling and the production of inflammatory cytokines by proalgesic agonists. Conditional deletion of Orai1 attenuated microglial proliferation in the dorsal horn, spinal cytokine levels, and potentiation of excitatory neurotransmission following peripheral nerve injury. These cellular effects were accompanied by mitigation of pain hyperalgesia in microglial Orai1 knockout mice. A small-molecule Orai1 inhibitor, CM4620, similarly mitigated allodynia in male mice. Unexpectedly, these protective effects were not seen in female mice, revealing sexual dimorphism in Orai1 regulation of microglial reactivity and hyperalgesia. Together, these findings indicate that Orai1 channels are key regulators of the sexually dimorphic role of microglia for the neuroinflammation that underlies neuropathic pain.


Asunto(s)
Microglía , Neuralgia , Ratones , Masculino , Femenino , Animales , Microglía/metabolismo , Hiperalgesia/genética , Enfermedades Neuroinflamatorias , Neuralgia/genética , Ratones Noqueados , Citocinas/metabolismo , Médula Espinal , Proteína ORAI1/genética
5.
EMBO Mol Med ; 14(9): e15687, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35919953

RESUMEN

Inflammatory bowel disease (IBD) is characterized by dysregulated intestinal immune responses. Using mass cytometry (CyTOF) to analyze the immune cell composition in the lamina propria (LP) of patients with ulcerative colitis (UC) and Crohn's disease (CD), we observed an enrichment of CD4+ effector T cells producing IL-17A and TNF, CD8+ T cells producing IFNγ, T regulatory (Treg) cells, and innate lymphoid cells (ILC). The function of these immune cells is regulated by store-operated Ca2+ entry (SOCE), which results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI and STIM proteins. We observed that the pharmacologic inhibition of SOCE attenuated the production of proinflammatory cytokines including IL-2, IL-4, IL-6, IL-17A, TNF, and IFNγ by human colonic T cells and ILCs, reduced the production of IL-6 by B cells and the production of IFNγ by myeloid cells, but had no effect on the viability, differentiation, and function of intestinal epithelial cells. T cell-specific deletion of CRAC channel genes in mice showed that Orai1, Stim1, and Stim2-deficient T cells have quantitatively distinct defects in SOCE, which correlate with gradually more pronounced impairment of cytokine production by Th1 and Th17 cells and the severity of IBD. Moreover, the pharmacologic inhibition of SOCE with a selective CRAC channel inhibitor attenuated IBD severity and colitogenic T cell function in mice. Our data indicate that SOCE inhibition may be a suitable new approach for the treatment of IBD.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio , Enfermedades Inflamatorias del Intestino , Animales , Linfocitos T CD8-positivos/metabolismo , Calcio/metabolismo , Canales de Calcio/metabolismo , Humanos , Inmunidad Innata , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Ratones , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/genética , Células Th17/metabolismo
6.
J Biol Chem ; 298(8): 102157, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35724962

RESUMEN

Stromal interaction molecule 1 (STIM1) is a widely expressed protein that functions as the endoplasmic reticulum (ER) Ca2+ sensor and activator of Orai1 channels. In resting cells with replete Ca2+ stores, an inhibitory clamp formed by the coiled-coil 1 (CC1) domain interacting with the CRAC-activation domain (CAD) of STIM1 helps keep STIM1 in a quiescent state. Following depletion of ER Ca2+ stores, the brake is released, allowing CAD to extend away from the ER membrane and enabling it to activate Orai1 channels. However, the molecular determinants of CC1-CAD interactions that enforce the inhibitory clamp are incompletely understood. Here, we performed Ala mutagenesis in conjunction with live-cell FRET analysis to examine residues in CC1 and CAD that regulate the inhibitory clamp. Our results indicate that in addition to previously identified hotspots in CC1⍺1 and CC3, several hydrophobic residues in CC2 and the apex region of CAD are critical for CC1-CAD interactions. Mutations in these residues loosen the CC1-CAD inhibitory clamp to release CAD from CC1 in cells with replete Ca2+ stores. By contrast, altering the hydrophobic residues L265 and L273 strengthens the clamp to prevent STIM1 activation. Inclusion of the inactivation domain of STIM1 helps stabilize CC1-CAD interaction in several mutants to prevent spontaneous STIM1 activation. In addition, R426C, a human disease-linked mutation in CC3, affects the clamp but also impairs Orai1 binding to inhibit CRAC channel activation. These results identify the CC2, apex, and inactivation domain regions of STIM1 as important determinants of STIM1 activation.


Asunto(s)
Señalización del Calcio , Retículo Endoplásmico , Molécula de Interacción Estromal 1 , Calcio/metabolismo , Canales de Calcio/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Dominios Proteicos , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
7.
Nat Commun ; 13(1): 2033, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440113

RESUMEN

TCR stimulation triggers Ca2+ signals that are critical for T cell function and immunity. Several pore-forming α and auxiliary ß subunits of voltage-gated Ca2+ channels (VGCC) were reported in T cells, but their mechanism of activation remains elusive and their contribution to Ca2+ signaling in T cells is controversial. We here identify CaVß1, encoded by Cacnb1, as a regulator of T cell function. Cacnb1 deletion enhances apoptosis and impairs the clonal expansion of T cells after lymphocytic choriomeningitis virus (LCMV) infection. By contrast, Cacnb1 is dispensable for T cell proliferation, cytokine production and Ca2+ signaling. Using patch clamp electrophysiology and Ca2+ recordings, we are unable to detect voltage-gated Ca2+ currents or Ca2+ influx in human and mouse T cells upon depolarization with or without prior TCR stimulation. mRNAs of several VGCC α1 subunits are detectable in human (CaV3.3, CaV3.2) and mouse (CaV2.1) T cells, but they lack transcription of many 5' exons, likely resulting in N-terminally truncated and non-functional proteins. Our findings demonstrate that although CaVß1 regulates T cell function, these effects are independent of VGCC channel activity.


Asunto(s)
Apoptosis , Linfocitos T , Animales , Apoptosis/genética , Canales de Calcio Tipo L , Proliferación Celular/genética , Ratones , Receptores de Antígenos de Linfocitos T
8.
Methods Enzymol ; 652: 213-239, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34059283

RESUMEN

Chemical modification of ion channels using the substituted cysteine accessibility method has a rich and successful history in elucidating the structural basis of ion channel function. In this approach, cysteine residues are introduced in regions of interest into the protein and their accessibility to water soluble thiol-reactive reagents is determined by monitoring ion channel activity. Because a wide range of these reagents are available with differing size, charge, and membrane solubility, the physio-chemical environment of the introduced cysteine residue and therefore the protein domain of interest can be probed with great precision. The approach has been widely employed for determining the secondary structure of specific ion channel domains, the location and nature of the channel gate, and the conformational rearrangements in the channel pore that underlie the opening/closing of the pore. In this chapter, we describe the use of these and related approaches to probe the functional architecture and gating of store-operated Orai1 channels.


Asunto(s)
Cisteína , Activación del Canal Iónico , Calcio/metabolismo , Proteína ORAI1/metabolismo , Dominios Proteicos
10.
Cell Calcium ; 94: 102366, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33581587

RESUMEN

Store-operated Orai channels are a primary mechanism for mobilizing Ca2+ signals in both non-excitable cells and excitable cells. The structure of the open channel, vital for understanding the mechanism of channel opening, is incompletely understood. We highlight a new study that unveils the structure of a constitutively active Orai mutant and takes us closer towards understanding the molecular basis of Orai channel activation.


Asunto(s)
Proteína ORAI1/metabolismo , Animales , Drosophila melanogaster/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Activación del Canal Iónico , Proteína ORAI1/química , Proteína ORAI1/ultraestructura , Conformación Proteica
11.
Cell Rep ; 33(9): 108464, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264616

RESUMEN

Store-operated Orai1 calcium channels function as highly Ca2+-selective ion channels and are broadly expressed in many tissues including the central nervous system, but their contributions to cognitive processing are largely unknown. Here, we report that many measures of synaptic, cellular, and behavioral models of learning are markedly attenuated in mice lacking Orai1 in forebrain excitatory neurons. Results with focal glutamate uncaging in hippocampal neurons support an essential role of Orai1 channels in amplifying NMDA-receptor-induced dendritic Ca2+ transients that drive activity-dependent spine morphogenesis and long-term potentiation at Schaffer collateral-CA1 synapses. Consistent with these signaling roles, mice lacking Orai1 in pyramidal neurons (but not interneurons) exhibit striking deficits in working and associative memory tasks. These findings identify Orai1 channels as essential regulators of dendritic spine Ca2+ signaling, synaptic plasticity, and cognition.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Espinas Dendríticas/metabolismo , Ácido Glutámico/metabolismo , Animales , Hipocampo/metabolismo , Memoria , Ratones , Proteína ORAI1 , Células Piramidales/metabolismo , Transducción de Señal
12.
Elife ; 92020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33124982

RESUMEN

Sulfur-aromatic interactions occur in the majority of protein structures, yet little is known about their functional roles in ion channels. Here, we describe a novel molecular motif, the M101 gate latch, which is essential for gating of human Orai1 channels via its sulfur-aromatic interactions with the F99 hydrophobic gate. Molecular dynamics simulations of different Orai variants reveal that the gate latch is mostly engaged in open but not closed channels. In experimental studies, we use metal-ion bridges to show that promoting an M101-F99 bond directly activates Orai1, whereas disrupting this interaction triggers channel closure. Mutational analysis demonstrates that the methionine residue at this position has a unique combination of length, flexibility, and chemistry to act as an effective latch for the phenylalanine gate. Because sulfur-aromatic interactions provide additional stabilization compared to purely hydrophobic interactions, we infer that the six M101-F99 pairs in the hexameric channel provide a substantial energetic contribution to Orai1 activation.


Asunto(s)
Activación del Canal Iónico/fisiología , Proteína ORAI1/metabolismo , Azufre/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Conformación Proteica , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Azufre/química
13.
J Physiol ; 598(23): 5391-5409, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32851638

RESUMEN

KEY POINTS: Temporal lobe epilepsy is a complex neurological disease caused by imbalance of excitation and inhibition in the brain. Growing literature implicates altered Ca2+ signalling in many aspects of epilepsy but the diversity of Ca2+ channels that regulate this syndrome are not well-understood. Here, we report that mice lacking the store-operated Ca2+ channel, Orai1, in the brain show markedly stronger seizures in response to the chemoconvulsants, kainic acid and pilocarpine. Electrophysiological analysis reveals that selective deletion of Orai1 channels in inhibitory neurons disables chemoconvulsant-induced excitation of GABAergic neurons in the CA1 hippocampus. Likewise, deletion of Orai1 in GABAergic neurons abrogates the chemoconvulsant-induced burst of spontaneous inhibitory postsynaptic currents (sIPSCs) on CA1 pyramidal neurons in the hippocampus. This loss of chemoconvulsant inhibition likely aggravates status epilepticus in Orai1 KO mice. These results identify Orai1 channels as regulators of hippocampal interneuron excitability and seizures. ABSTRACT: Store-operated Orai1 channels are a major mechanism for Ca2+ entry in many cells and mediate numerous functions including gene expression, cytokine production and gliotransmitter release. Orai1 is expressed in many regions of the mammalian brain; however, its role in regulating neuronal excitability, synaptic function and brain disorders has only now begun to be investigated. To investigate a potential role of Orai1 channels in status epilepticus induced by chemoconvulsants, we examined acute seizures evoked by intraperitoneal injections of kainic acid (KA) and pilocarpine in mice with a conditional deletion of Orai1 (or its activator STIM1) in the brain. Brain-specific Orai1 and STIM1 knockout (KO) mice exhibited significantly stronger seizures (P = 0.00003 and P < 0.00001), and higher chemoconvulsant-induced mortality (P = 0.02) compared with wildtype (WT) littermates. Electrophysiological recordings in hippocampal brain slices revealed that KA stimulated the activity of inhibitory interneurons in the CA1 hippocampus (P = 0.04) which failed to occur in Orai1 KO mice. Further, KA and pilocarpine increased the frequency of spontaneous IPSCs in CA1 pyramidal neurons >twofold (KA: P = 0.04; pilocarpine: P = 0.0002) which was abolished in Orai1 KO mice. Mice with selective deletion of Orai1 in GABAergic neurons alone also showed stronger seizures to KA (P = 0.001) and pilocarpine (P < 0.00001) and loss of chemoconvulsant-induced increases in sIPSC responses compared with WT controls. We conclude that Orai1 channels regulate chemoconvulsant-induced excitation in GABAergic neurons and that destabilization of the excitatory/inhibitory balance in Orai1 KO mice aggravates chemoconvulsant-mediated seizures. These results identify Orai1 channels as novel molecular regulators of hippocampal neuronal excitability and seizures.


Asunto(s)
Hipocampo , Convulsiones , Animales , Ácido Kaínico/toxicidad , Ratones , Proteína ORAI1/genética , Pilocarpina/toxicidad , Células Piramidales , Convulsiones/inducido químicamente
14.
J Vet Med Sci ; 82(5): 646-652, 2020 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-32213732

RESUMEN

The effects of climatic elements on Salmonella contamination of chicken meat were investigated. Logistic regression analysis was performed to evaluate the association between Salmonella isolation, for 240 chicken samples purchased from March 2015 to February 2017, and climatic elements, over 65 days of chicken rearing. Salmonella was isolated from 143 samples (59.6%), and the most dominant serovars identified were Infantis (77/240, 32.1%) and Schwarzengrund (56/240, 23.3%). Previous studies have reported S. Schwarzengrund contamination of broiler chickens only in western Japan; however, in the present study, S. Schwarzengrund was also isolated from meat produced in eastern Japan-20% (12/60) in the C prefecture to 36.4% (8/22) in the Y prefecture-suggesting that S. Schwarzengrund-contaminated areas have expanded towards eastern Japan. Air temperature showed a significant negative association with S. Schwarzengrund isolation for chicken meat produced during periods with rising temperature (spring and summer) [odds ratio (OR), 0.894 to 0.935; P<0.01]. Moreover, the risk of S. Schwarzengrund contamination of chicken meat was higher during spring (OR, 3.951; P=0.008) and winter (OR, 4.071; P=0.006) than during summer. Effects of climatic elements and differences in contamination risk across seasons were not observed for any Salmonella serovars and only S. Infantis, which could be attributed to differences in transmission patterns and vehicles among Salmonella serovars. These findings are valuable for understanding the dynamics of S. Schwarzengrund dissemination in broiler farms.


Asunto(s)
Microbiología de Alimentos , Productos Avícolas/microbiología , Salmonella/aislamiento & purificación , Animales , Pollos , Clima , Contaminación de Alimentos/análisis , Japón , Salmonella/clasificación , Estaciones del Año , Serogrupo
15.
J Physiol ; 598(9): 1707-1723, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-30950063

RESUMEN

Store-operated Ca2+ entry through Orai1 channels is a primary mechanism for Ca2+ entry in many cells and mediates numerous cellular effector functions ranging from gene transcription to exocytosis. Orai1 channels are amongst the most Ca2+ -selective channels known and are activated by direct physical interactions with the endoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1) in response to store depletion triggered by stimulation of a variety of cell surface G-protein coupled and tyrosine kinase receptors. Work in the last decade has revealed that the Orai1 gating process is highly cooperative and strongly allosteric, likely driven by a wave of interdependent conformational changes throughout the protein originating in the peripheral C-terminal ligand binding site and culminating in pore opening. In this review, we survey the structural and molecular features in Orai1 that contribute to channel gating and consider how they give rise to the unique biophysical fingerprint of Orai1 currents.


Asunto(s)
Canales de Calcio , Activación del Canal Iónico , Calcio/metabolismo , Canales de Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Proteína ORAI1 , Molécula de Interacción Estromal 1
16.
J Gen Physiol ; 152(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31816637

RESUMEN

Store-operated Orai1 channels regulate a wide range of cellular functions from gene expression to cell proliferation. Previous studies have shown that gating of Orai1 channels is regulated by the outer pore residues V102 and F99, which together function as a hydrophobic gate to block ion conduction in resting channels. Opening of this gate occurs through a conformational change that moves F99 away from the permeation pathway, leading to pore hydration and ion conduction. In addition to this outer hydrophobic gate, several studies have postulated the presence of an inner gate formed by the basic residues R91, K87, and R83 in the inner pore. These positively charged residues were suggested to block ion conduction in closed channels via mechanisms involving either electrostatic repulsion or steric occlusion by a bound anion plug. However, in contrast to this model, here we find that neutralization of the basic residues dose-dependently abolishes both STIM1-mediated and STIM1-independent activation of Orai1 channels. Molecular dynamics simulations show that loss of the basic residues dehydrates the pore around the hydrophobic gate and stabilizes the pore in a closed configuration. Likewise, the severe combined immunodeficiency mutation, Orai1 R91W, closes the channel by dewetting the hydrophobic stretch of the pore and stabilizing F99 in a pore-facing configuration. Loss of STIM1-gating in R91W and in the other basic residue mutants is rescued by a V102A mutation, which restores pore hydration at the hydrophobic gate to repermit ion conduction. These results indicate that the inner pore basic residues facilitate opening of the principal outer hydrophobic gate through a long-range effect involving hydration of the outer pore.


Asunto(s)
Sustitución de Aminoácidos , Activación del Canal Iónico , Proteína ORAI1/química , Arginina/química , Arginina/genética , Células HEK293 , Humanos , Lisina/química , Lisina/genética , Simulación de Dinámica Molecular , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Dominios Proteicos
17.
Proc Natl Acad Sci U S A ; 115(22): E5193-E5202, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29760086

RESUMEN

Store-operated Orai1 channels are activated through a unique inside-out mechanism involving binding of the endoplasmic reticulum Ca2+ sensor STIM1 to cytoplasmic sites on Orai1. Although atomic-level details of Orai structure, including the pore and putative ligand binding domains, are resolved, how the gating signal is communicated to the pore and opens the gate is unknown. To address this issue, we used scanning mutagenesis to identify 15 residues in transmembrane domains (TMs) 1-4 whose perturbation activates Orai1 channels independently of STIM1. Cysteine accessibility analysis and molecular-dynamics simulations indicated that constitutive activation of the most robust variant, H134S, arises from a pore conformational change that opens a hydrophobic gate to augment pore hydration, similar to gating evoked by STIM1. Mutational analysis of this locus suggests that H134 acts as steric brake to stabilize the closed state of the channel. In addition, atomic packing analysis revealed distinct functional contacts between the TM1 pore helix and the surrounding TM2/3 helices, including one set mediated by a cluster of interdigitating hydrophobic residues and another by alternative ridges of polar and hydrophobic residues. Perturbing these contacts via mutagenesis destabilizes STIM1-mediated Orai1 channel gating, indicating that these bridges between TM1 and the surrounding TM2/3 ring are critical for conveying the gating signal to the pore. These findings help develop a framework for understanding the global conformational changes and allosteric interactions between topologically distinct domains that are essential for activation of Orai1 channels.


Asunto(s)
Calcio/química , Calcio/metabolismo , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Humanos , Simulación de Dinámica Molecular , Proteína ORAI1/genética , Dominios Proteicos
18.
Nat Commun ; 8: 14714, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294127

RESUMEN

Store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels is critical for lymphocyte function and immune responses. CRAC channels are hexamers of ORAI proteins that form the channel pore, but the contributions of individual ORAI homologues to CRAC channel function are not well understood. Here we show that deletion of Orai1 reduces, whereas deletion of Orai2 increases, SOCE in mouse T cells. These distinct effects are due to the ability of ORAI2 to form heteromeric channels with ORAI1 and to attenuate CRAC channel function. The combined deletion of Orai1 and Orai2 abolishes SOCE and strongly impairs T cell function. In vivo, Orai1/Orai2 double-deficient mice have impaired T cell-dependent antiviral immune responses, and are protected from T cell-mediated autoimmunity and alloimmunity in models of colitis and graft-versus-host disease. Our study demonstrates that ORAI1 and ORAI2 form heteromeric CRAC channels, in which ORAI2 fine-tunes the magnitude of SOCE to modulate immune responses.


Asunto(s)
Calcio/metabolismo , Inmunidad , Proteína ORAI2/metabolismo , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Proliferación Celular , Colitis/inmunología , Colitis/patología , Citocinas/biosíntesis , Eliminación de Gen , Regulación de la Expresión Génica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Homeostasis , Humanos , Inmunidad Humoral , Activación del Canal Iónico , Recuento de Linfocitos , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Proteína ORAI1/deficiencia , Proteína ORAI1/metabolismo , Proteína ORAI2/deficiencia , Multimerización de Proteína , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo
19.
Nat Commun ; 8: 14512, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28220789

RESUMEN

Store-operated Ca2+ release-activated Ca2+ (CRAC) channels constitute a major pathway for Ca2+ influx and mediate many essential signalling functions in animal cells, yet how they open remains elusive. Here, we investigate the gating mechanism of the human CRAC channel Orai1 by its activator, stromal interacting molecule 1 (STIM1). We find that two rings of pore-lining residues, V102 and F99, work together to form a hydrophobic gate. Mutations of these residues to polar amino acids produce channels with leaky gates that conduct ions in the resting state. STIM1-mediated channel activation occurs through rotation of the pore helix, which displaces the F99 residues away from the pore axis to increase pore hydration, allowing ions to flow through the V102-F99 hydrophobic band. Pore helix rotation by STIM1 also explains the dynamic coupling between CRAC channel gating and ion selectivity. This hydrophobic gating mechanism has implications for CRAC channel function, pharmacology and disease-causing mutations.


Asunto(s)
Calcio/metabolismo , Activación del Canal Iónico , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Microscopía Confocal , Modelos Moleculares , Mutación , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteína ORAI1/química , Proteína ORAI1/genética , Fenilalanina/química , Fenilalanina/genética , Fenilalanina/metabolismo , Estructura Secundaria de Proteína , Rotación , Molécula de Interacción Estromal 1/química , Molécula de Interacción Estromal 1/genética , Valina/química , Valina/genética , Valina/metabolismo
20.
Cell Calcium ; 63: 14-19, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28108030

RESUMEN

Three decades ago, James W. Putney Jr. conceptualized the idea of store-operated calcium entry (SOCE) to explain how depletion of endoplasmic reticulum (ER) Ca2+ stores evokes Ca2+ influx across the plasma membrane. Since the publication of this highly influential idea, it is now established that SOCE is universal among non-excitable and probably even many types of excitable cells, and contributes to numerous effector functions impacting immunity, muscle contraction, and brain function. The molecules encoding SOCE, the STIM and Orai proteins, are now known and our understanding of how this pathway is activated in response to ER Ca2+ store depletion has advanced significantly. In this review, we summarize the current knowledge of how Orai1 channels are activated by STIM1, focusing on recent work supporting a hydrophobic gating mechanism for the opening of the Orai1 channel pore.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...