Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Environ Monit Assess ; 196(7): 616, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38874785

RESUMO

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.


Assuntos
Monitoramento Ambiental , Florestas , Animais , Monitoramento Ambiental/métodos , Japão , Imagens de Satélites , Tecnologia de Sensoriamento Remoto , Ecossistema , Mariposas/fisiologia , Larva
2.
J Biol Chem ; 298(8): 102157, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35724962

RESUMO

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.


Assuntos
Sinalização do Cálcio , Retículo Endoplasmático , Molécula 1 de Interação Estromal , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Domínios Proteicos , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(22): E5193-E5202, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29760086

RESUMO

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.


Assuntos
Cálcio/química , Cálcio/metabolismo , Proteína ORAI1/química , Proteína ORAI1/metabolismo , Humanos , Simulação de Dinâmica Molecular , Proteína ORAI1/genética , Domínios Proteicos
4.
J Physiol ; 598(9): 1707-1723, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-30950063

RESUMO

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.


Assuntos
Canais de Cálcio , Ativação do Canal Iônico , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Proteína ORAI1 , Molécula 1 de Interação Estromal
5.
J Physiol ; 598(23): 5391-5409, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32851638

RESUMO

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.


Assuntos
Hipocampo , Convulsões , Animais , Ácido Caínico/toxicidade , Camundongos , Proteína ORAI1/genética , Pilocarpina/toxicidade , Células Piramidais , Convulsões/induzido quimicamente
6.
Nat Immunol ; 9(4): 432-43, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18327260

RESUMO

Store-operated Ca2+ entry through calcium release-activated calcium channels is the chief mechanism for increasing intracellular Ca2+ in immune cells. Here we show that mouse T cells and fibroblasts lacking the calcium sensor STIM1 had severely impaired store-operated Ca2+ influx, whereas deficiency in the calcium sensor STIM2 had a smaller effect. However, T cells lacking either STIM1 or STIM2 had much less cytokine production and nuclear translocation of the transcription factor NFAT. T cell-specific ablation of both STIM1 and STIM2 resulted in a notable lymphoproliferative phenotype and a selective decrease in regulatory T cell numbers. We conclude that both STIM1 and STIM2 promote store-operated Ca2+ entry into T cells and fibroblasts and that STIM proteins are required for the development and function of regulatory T cells.


Assuntos
Retículo Endoplasmático/metabolismo , Tolerância Imunológica , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/fisiologia , Linfócitos T Reguladores/imunologia , Sequência de Aminoácidos , Animais , Transporte Biológico Ativo/genética , Transporte Biológico Ativo/imunologia , Cálcio/metabolismo , Canais de Cálcio , Linhagem Celular , Linhagem Celular Transformada , Células Cultivadas , Retículo Endoplasmático/fisiologia , Humanos , Tolerância Imunológica/genética , Ativação Linfocitária/genética , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal
8.
Nature ; 482(7384): 241-5, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22278058

RESUMO

Two defining functional features of ion channels are ion selectivity and channel gating. Ion selectivity is generally considered an immutable property of the open channel structure, whereas gating involves transitions between open and closed channel states, typically without changes in ion selectivity. In store-operated Ca(2+) release-activated Ca(2+) (CRAC) channels, the molecular mechanism of channel gating by the CRAC channel activator, stromal interaction molecule 1 (STIM1), remains unknown. CRAC channels are distinguished by a very high Ca(2+) selectivity and are instrumental in generating sustained intracellular calcium concentration elevations that are necessary for gene expression and effector function in many eukaryotic cells. Here we probe the central features of the STIM1 gating mechanism in the human CRAC channel protein, ORAI1, and identify V102, a residue located in the extracellular region of the pore, as a candidate for the channel gate. Mutations at V102 produce constitutively active CRAC channels that are open even in the absence of STIM1. Unexpectedly, although STIM1-free V102 mutant channels are not Ca(2+)-selective, their Ca(2+) selectivity is dose-dependently boosted by interactions with STIM1. Similar enhancement of Ca(2+) selectivity is also seen in wild-type ORAI1 channels by increasing the number of STIM1 activation domains that are directly tethered to ORAI1 channels, or by increasing the relative expression of full-length STIM1. Thus, exquisite Ca(2+) selectivity is not an intrinsic property of CRAC channels but rather a tuneable feature that is bestowed on otherwise non-selective ORAI1 channels by STIM1. Our results demonstrate that STIM1-mediated gating of CRAC channels occurs through an unusual mechanism in which permeation and gating are closely coupled.


Assuntos
Canais de Cálcio/metabolismo , Ativação do Canal Iônico , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canais de Cálcio/química , Canais de Cálcio/genética , Células HEK293 , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Modelos Moleculares , Mutação/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteína ORAI1 , Molécula 1 de Interação Estromal , Relação Estrutura-Atividade
9.
J Immunol ; 195(5): 2122-33, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26238490

RESUMO

The G-protein-coupled protease-activated receptor 2 (PAR2) plays an important role in the pathogenesis of various inflammatory and auto-immune disorders. In airway epithelial cells (AECs), stimulation of PAR2 by allergens and proteases triggers the release of a host of inflammatory mediators to regulate bronchomotor tone and immune cell recruitment. Activation of PAR2 turns on several cell signaling pathways of which the mobilization of cytosolic Ca(2+) is likely a critical but poorly understood event. In this study, we show that Ca(2+) release-activated Ca(2+) (CRAC) channels encoded by stromal interaction molecule 1 and Orai1 are a major route of Ca(2+) entry in primary human AECs and drive the Ca(2+) elevations seen in response to PAR2 activation. Activation of CRAC channels induces the production of several key inflammatory mediators from AECs including thymic stromal lymphopoietin, IL-6, and PGE2, in part through stimulation of gene expression via nuclear factor of activated T cells (NFAT). Furthermore, PAR2 stimulation induces the production of many key inflammatory mediators including PGE2, IL-6, IL-8, and GM-CSF in a CRAC channel-dependent manner. These findings indicate that CRAC channels are the primary mechanism for Ca(2+) influx in AECs and a vital checkpoint for the induction of PAR2-induced proinflammatory cytokines.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Citocinas/metabolismo , Células Epiteliais/metabolismo , Receptor PAR-2/metabolismo , Western Blotting , Brônquios/citologia , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Retículo Endoplasmático/metabolismo , Células Epiteliais/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Interferência de RNA , Receptor PAR-2/agonistas , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal , Tripsina/metabolismo
10.
Oecologia ; 177(1): 191-202, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25362582

RESUMO

Knowledge of variations in morphophysiological leaf traits with forest height is essential for quantifying carbon and water fluxes from forest ecosystems. Here, we examined changes in leaf traits with forest height in diverse tree species and their role in environmental acclimation in a tropical rain forest in Borneo that does not experience dry spells. Height-related changes in leaf physiological and morphological traits [e.g., maximum photosynthetic rate (Amax), stomatal conductance (gs), dark respiration rate (Rd), carbon isotope ratio (δ(13)C), nitrogen (N) content, and leaf mass per area (LMA)] from understory to emergent trees were investigated in 104 species in 29 families. We found that many leaf area-based physiological traits (e.g., A(max-area), Rd, gs), N, δ(13)C, and LMA increased linearly with tree height, while leaf mass-based physiological traits (e.g., A(max-mass)) only increased slightly. These patterns differed from other biomes such as temperate and tropical dry forests, where trees usually show decreased photosynthetic capacity (e.g., A(max-area), A(max-mass)) with height. Increases in photosynthetic capacity, LMA, and δ(13)C are favored under bright and dry upper canopy conditions with higher photosynthetic productivity and drought tolerance, whereas lower R d and LMA may improve shade tolerance in lower canopy trees. Rapid recovery of leaf midday water potential to theoretical gravity potential during the night supports the idea that the majority of trees do not suffer from strong drought stress. Overall, leaf area-based photosynthetic traits were associated with tree height and the degree of leaf drought stress, even in diverse tropical rain forest trees.


Assuntos
Aclimatação , Florestas , Fotossíntese , Folhas de Planta/fisiologia , Árvores/fisiologia , Clima Tropical , Bornéu , Carbono/metabolismo , Isótopos de Carbono/metabolismo , Nitrogênio/metabolismo , Folhas de Planta/crescimento & desenvolvimento , Chuva , Árvores/crescimento & desenvolvimento
11.
Artigo em Inglês | MEDLINE | ID: mdl-38836042

RESUMO

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.

12.
J Physiol ; 591(11): 2833-50, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23613525

RESUMO

Ca(2+) release-activated Ca(2+) (CRAC) channels are activated through a mechanism wherein depletion of intracellular calcium stores results in the aggregation of stromal interaction molecule 1 (STIM1), the endoplasmic reticulum (ER) Ca(2+) sensor, and Orai1, the CRAC channel protein, at overlapping sites in the ER and plasma membranes (PMs). The redistribution of CRAC channels is driven through direct STIM1-Orai1 binding, an important event that not only controls gating, but also regulates Orai1 ion selectivity. Orai1 harbours two STIM1 binding sites, one each on the intracellular C- and N-termini. Previous studies have proposed modular functions for these sites, with the C-terminal site thought to regulate STIM1-Orai1 binding and trapping of Orai1 at the ER-PM junctions, and the N-terminal site mediating gating. However, here we find that a variety of mutations in the N-terminal site impair the binding of Orai1 to STIM1 and to the soluble CRAC activation domain (CAD). Gating could be restored in several N- and C-terminal point mutants by directly tethering the minimal STIM1 activation domain (S) to Orai1 (Orai1-SS channels), indicating that loss of gating in these mutants by full-length STIM1 results from insufficient ligand binding. By contrast, gating could not be restored in mutant Orai1-SS channels carrying more drastic deletions that removed the STIM1 binding sites (1-85, 73-85, or 272-279 Orai1), suggesting that STIM1 binding to both sites is essential for channel activation. Moreover, analysis of ion selectivity indicated that the molecular requirements for gating and modulation of ion selectivity are similar, yet substantively different from those for Orai1 puncta formation, suggesting that ion selectivity and gating are mechanistically coupled in CRAC channels. Our results indicate that the C- and N-terminal STIM1 binding sites are both essential for multiple aspects of Orai1 function including STIM1-Orai1 association, Orai1 trapping, and channel activation.


Assuntos
Canais de Cálcio/metabolismo , Ativação do Canal Iônico , Glicoproteínas de Membrana/metabolismo , Subunidades Proteicas/metabolismo , Sítios de Ligação , Canais de Cálcio/química , Canais de Cálcio/genética , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Deleção de Genes , Células HEK293 , Humanos , Mutação Puntual , Ligação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Transporte Proteico
13.
Elife ; 122023 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-36806330

RESUMO

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.


Assuntos
Canais de Cálcio , Canais de Cálcio Ativados pela Liberação de Cálcio , Humanos , Canais de Cálcio/metabolismo , Proteína ORAI1/genética , Mutação , Canais de Cálcio Ativados pela Liberação de Cálcio/genética , Mutação com Ganho de Função , Molécula 1 de Interação Estromal/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
14.
Sci Adv ; 9(4): eade7002, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36706180

RESUMO

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.


Assuntos
Microglia , Neuralgia , Camundongos , Masculino , Feminino , Animais , Microglia/metabolismo , Hiperalgesia/genética , Doenças Neuroinflamatórias , Neuralgia/genética , Camundongos Knockout , Citocinas/metabolismo , Medula Espinal , Proteína ORAI1/genética
15.
J Biol Chem ; 286(11): 9429-42, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21193399

RESUMO

Activation of Ca(2+) release-activated Ca(2+) channels by depletion of intracellular Ca(2+) stores involves physical interactions between the endoplasmic reticulum Ca(2+) sensor, STIM1, and the channels composed of Orai subunits. Recent studies indicate that the Orai3 subtype, in addition to being store-operated, is also activated in a store-independent manner by 2-aminoethyldiphenyl borate (2-APB), a small molecule with complex pharmacology. However, it is unknown whether the store-dependent and -independent activation modes of Orai3 channels operate independently or whether there is cross-talk between these activation states. Here we report that in addition to causing direct activation, 2-APB also regulates store-operated gating of Orai3 channels, causing potentiation at low doses and inhibition at high doses. Inhibition of store-operated gating by 2-APB was accompanied by the suppression of several modes of Orai3 channel regulation that depend on STIM1, suggesting that high doses of 2-APB interrupt STIM1-Orai3 coupling. Conversely, STIM1-bound Orai3 (and Orai1) channels resisted direct gating by high doses of 2-APB. The rate of direct 2-APB activation of Orai3 channels increased linearly with the degree of STIM1-Orai3 uncoupling, suggesting that 2-APB has to first disengage STIM1 before it can directly gate Orai3 channels. Collectively, our results indicate that the store-dependent and -independent modes of Ca(2+) release-activated Ca(2+) channel activation are mutually exclusive: channels bound to STIM1 resist 2-APB gating, whereas 2-APB antagonizes STIM1 gating.


Assuntos
Compostos de Boro/farmacologia , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Proteína ORAI1 , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Molécula 1 de Interação Estromal
16.
J Immunol ; 185(10): 5845-58, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20956344

RESUMO

ORAI1 is the pore-forming subunit of the Ca(2+) release-activated Ca(2+) (CRAC) channel, which is responsible for store-operated Ca(2+) entry in lymphocytes. A role for ORAI1 in T cell function in vivo has been inferred from in vitro studies of T cells from human immunodeficient patients with mutations in ORAI1 and Orai1(-/-) mice, but a detailed analysis of T cell-mediated immune responses in vivo in mice lacking functional ORAI1 has been missing. We therefore generated Orai1 knock-in mice (Orai1(KI/KI)) expressing a nonfunctional ORAI1-R93W protein. Homozygosity for the equivalent ORAI1-R91W mutation abolishes CRAC channel function in human T cells resulting in severe immunodeficiency. Homozygous Orai1(KI/KI) mice die neonatally, but Orai1(KI/KI) fetal liver chimeric mice are viable and show normal lymphocyte development. T and B cells from Orai1(KI/KI) mice display severely impaired store-operated Ca(2+) entry and CRAC channel function resulting in a strongly reduced expression of several key cytokines including IL-2, IL-4, IL-17, IFN-γ, and TNF-α in CD4(+) and CD8(+) T cells. Cell-mediated immune responses in vivo that depend on Th1, Th2, and Th17 cell function were severely attenuated in ORAI1-deficient mice. Orai1(KI/KI) mice lacked detectable contact hypersensitivity responses and tolerated skin allografts significantly longer than wild-type mice. In addition, T cells from Orai1(KI/KI) mice failed to induce colitis in an adoptive transfer model of inflammatory bowel disease. These findings reaffirm the critical role of ORAI1 for T cell function and provide important insights into the in vivo functions of CRAC channels for T cell-mediated immunity.


Assuntos
Autoimunidade/imunologia , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Rejeição de Enxerto/metabolismo , Linfócitos T/imunologia , Animais , Canais de Cálcio/genética , Canais de Cálcio/imunologia , Sinalização do Cálcio/genética , Sinalização do Cálcio/imunologia , Separação Celular , Citocinas/biossíntese , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Técnicas de Introdução de Genes , Rejeição de Enxerto/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Mutação , Proteína ORAI1 , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Quimeras de Transplante , Transplante Homólogo
17.
Proc Natl Acad Sci U S A ; 106(52): 22516-21, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20018736

RESUMO

CRAC channels generate Ca(2+) signals critical for the activation of immune cells and exhibit an intriguing pore profile distinguished by extremely high Ca(2+) selectivity, low Cs(+) permeability, and small unitary conductance. To identify the ion conduction pathway and gain insight into the structural bases of these permeation characteristics, we introduced cysteine residues in the CRAC channel pore subunit, Orai1, and probed their accessibility to various thiol-reactive reagents. Our results indicate that the architecture of the ion conduction pathway is characterized by a flexible outer vestibule formed by the TM1-TM2 loop, which leads to a narrow pore flanked by residues of a helical TM1 segment. Residues in TM3, and specifically, E190, a residue considered important for ion selectivity, are not close to the pore. Moreover, the outer vestibule does not significantly contribute to ion selectivity, implying that Ca(2+) selectivity is conferred mainly by E106. The ion conduction pathway is sufficiently narrow along much of its length to permit stable coordination of Cd(2+) by several TM1 residues, which likely explains the slow flux of ions within the restrained geometry of the pore. These results provide a structural framework to understand the unique permeation properties of CRAC channels.


Assuntos
Canais de Cálcio/química , Canais de Cálcio/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Cálcio/metabolismo , Canais de Cálcio/genética , Cisteína/química , Humanos , Técnicas In Vitro , Transporte de Íons , Lantânio/metabolismo , Mesilatos/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteína ORAI1 , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Reagentes de Sulfidrila/farmacologia
18.
Nat Commun ; 13(1): 2033, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35440113

RESUMO

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.


Assuntos
Apoptose , Linfócitos T , Animais , Apoptose/genética , Canais de Cálcio Tipo L , Proliferação de Células/genética , Camundongos , Receptores de Antígenos de Linfócitos T
19.
EMBO Mol Med ; 14(9): e15687, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35919953

RESUMO

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.


Assuntos
Canais de Cálcio Ativados pela Liberação de Cálcio , Doenças Inflamatórias Intestinais , Animais , Linfócitos T CD8-Positivos/metabolismo , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Humanos , Imunidade Inata , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Camundongos , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/genética , Células Th17/metabolismo
20.
Methods Enzymol ; 652: 213-239, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34059283

RESUMO

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.


Assuntos
Cisteína , Ativação do Canal Iônico , Cálcio/metabolismo , Proteína ORAI1/metabolismo , Domínios Proteicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA