Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Oncogene ; 42(24): 1980-1993, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37130916

RESUMEN

Peritoneal metastasis (PM) is an important metastatic modality of gastric cancer (GC).It is associated with poor prognosis. The underlying molecular mechanism of PM remains elusive. 5-Methylcytosine (m5C), a posttranscriptional RNA modification, involves in the progression of many tumors. However, its role in GC peritoneal metastasis remains unclear. In our study, transcriptome results suggested that NSUN2 expression was significantly upregulated in PM. And patients with high NSUN2 expression of PM predicted a worse prognosis. Mechanistically, NSUN2 regulates ORAI2 mRNA stability by m5C modification, thereby promoting ORAI2 expression and further promoting peritoneal metastasis and colonization of GC. YBX1 acts as a "reader" by binding to the ORAI2 m5C modification site. Following the uptake of fatty acids from omental adipocytes by GC cells, the transcription factor E2F1 was upregulated, which further promoted the expression of NSUN2 through cis-element. Briefly, these results revealed that peritoneal adipocytes provide fatty acid for GC cells, thus contributing to the elevation of E2F1 and NSUN2 through AMPK pathway, and upregulated NSUN2 activates the key gene ORAI2 through m5C modification, thereby promoting peritoneal metastasis and colonization of gastric cancer.


Asunto(s)
Neoplasias Peritoneales , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Metiltransferasas/metabolismo , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/secundario , Procesamiento Postranscripcional del ARN , Proteína ORAI2/genética , Proteína ORAI2/metabolismo
2.
Glia ; 70(9): 1666-1680, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35506586

RESUMEN

Astrocytes are glial cells that serve homeostatic functions in the central nervous system (CNS). Recent research, however, suggests that under pathological conditions, astrocytes are stimulated by various factors and actively participate in CNS inflammation. In the present study, we found that astrocytes upregulate various inflammatory factors including prostaglandin E2 (PGE2 ) by co-stimulation with tumor necrosis factor-alpha (TNFα) and interleukin-1alpha (IL1α). These TNFα/IL1α-stimulated astrocytes also showed increased Ca2+ release from the endoplasmic reticulum (ER) and increased expression of Orai2, a member of the store-operated calcium channel (SOCC) family. To reveal the role of Orai2, we used astrocytes in which Orai2 was knocked-down (KD) or knocked-out (KO). The expression of the prostaglandin E synthase Ptges and the production of PGE2 were higher in Orai2-KD astrocytes than in WT astrocytes when stimulated with TNFα and IL1α. Orai2-KO astrocytes also showed increased expression of Ptges and increased PGE2 production. The expression of Ptgs2, another PGE2 synthetic enzyme, was also upregulated in Orai2-KO astrocytes. Moreover, Orai2-KO astrocytes showed increased store-operated calcium entry (SOCE) and increased Orai1 expression. These results suggest that Orai2 is upregulated in TNFα/IL1α-stimulated astrocytes and reduces PGE2 production to some extent, modulating CNS inflammation. Our findings may aid in understanding how astrocytes are associated with inflammatory responses, and the identification of new targets that modulate astrocytic reactivity.


Asunto(s)
Astrocitos , Interleucina-1alfa , Proteína ORAI2 , Prostaglandinas E , Factor de Necrosis Tumoral alfa , Animales , Astrocitos/metabolismo , Calcio/metabolismo , Señalización del Calcio , Inflamación , Interleucina-1alfa/metabolismo , Interleucina-1alfa/farmacología , Ratones , Proteína ORAI2/metabolismo , Prostaglandinas E/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
3.
J Physiol ; 600(3): 623-643, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34877682

RESUMEN

Three Orai (Orai1, Orai2, and Orai3) and two stromal interaction molecule (STIM1 and STIM2) mammalian protein homologues constitute major components of the store-operated Ca2+ entry mechanism. When co-expressed with STIM1, Orai1, Orai2 and Orai3 form highly selective Ca2+ channels with properties of Ca2+ release-activated Ca2+ (CRAC) channels. Despite the high level of homology between Orai proteins, CRAC channels formed by different Orai isoforms have distinctive properties, particularly with regards to Ca2+ -dependent inactivation, inhibition/potentiation by 2-aminoethyl diphenylborinate and sensitivity to reactive oxygen species. This study characterises and compares the regulation of Orai1, Orai2- and Orai3-mediated CRAC current (ICRAC ) by intracellular pH (pHi ). Using whole-cell patch clamping of HEK293T cells heterologously expressing Orai and STIM1, we show that ICRAC formed by each Orai homologue has a unique sensitivity to changes in pHi . Orai1-mediated ICRAC exhibits a strong dependence on pHi of both current amplitude and the kinetics of Ca2+ -dependent inactivation. In contrast, Orai2 amplitude, but not kinetics, depends on pHi , whereas Orai3 shows no dependence on pHi at all. Investigation of different Orai1-Orai3 chimeras suggests that pHi dependence of Orai1 resides in both the N-terminus and intracellular loop 2, and may also involve pH-dependent interactions with STIM1. KEY POINTS: It has been shown previously that Orai1/stromal interaction molecule 1 (STIM1)-mediated Ca2+ release-activated Ca2+ current (ICRAC ) is inhibited by intracellular acidification and potentiated by intracellular alkalinisation. The present study reveals that CRAC channels formed by each of the Orai homologues Orai1, Orai2 and Orai3 has a unique sensitivity to changes in intracellular pH (pHi ). The amplitude of Orai2 current is affected by the changes in pHi  similarly to the amplitude of Orai1. However, unlike Orai1, fast Ca2+ -dependent inactivation of Orai2 is unaffected by acidic pHi . In contrast to both Orai1 and Orai2, Orai3 is not sensitive to pHi  changes. Domain swapping between Orai1 and Orai3 identified the N-terminus and intracellular loop 2 as the molecular structures responsible for Orai1 regulation by pHi . Reduction of ICRAC dependence on pHi seen in a STIM1-independent Orai1 mutant suggested that some parts of STIM1 are also involved in ICRAC modulation by pHi .


Asunto(s)
Canales de Calcio , Canales de Calcio Activados por la Liberación de Calcio , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Proteína ORAI1/genética , Proteína ORAI2/metabolismo , Molécula de Interacción Estromal 1/metabolismo
4.
Nat Commun ; 12(1): 7035, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857756

RESUMEN

RNA editing is a feature of RNA maturation resulting in the formation of transcripts whose sequence differs from the genome template. Brain RNA editing may be altered in Alzheimer's disease (AD). Here, we analyzed data from 1,865 brain samples covering 9 brain regions from 1,074 unrelated subjects on a transcriptome-wide scale to identify inter-regional differences in RNA editing. We expand the list of known brain editing events by identifying 58,761 previously unreported events. We note that only a small proportion of these editing events are found at the protein level in our proteome-wide validation effort. We also identified the occurrence of editing events associated with AD dementia, neuropathological measures and longitudinal cognitive decline in: SYT11, MCUR1, SOD2, ORAI2, HSDL2, PFKP, and GPRC5B. Thus, we present an extended reference set of brain RNA editing events, identify a subset that are found to be expressed at the protein level, and extend the narrative of transcriptomic perturbation in AD to RNA editing.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteína ORAI2/genética , Edición de ARN , ARN/genética , Sinaptotagminas/genética , Transcriptoma , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Atlas como Asunto , Encéfalo/metabolismo , Encéfalo/patología , Química Encefálica , Perfilación de la Expresión Génica , Humanos , Hidroxiesteroide Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteína ORAI2/metabolismo , Fosfofructoquinasa-1 Tipo C/genética , Fosfofructoquinasa-1 Tipo C/metabolismo , ARN/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Sinaptotagminas/metabolismo
5.
Int J Mol Sci ; 22(24)2021 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-34948414

RESUMEN

Store-operated calcium entry (SOCE) constitutes a fine-tuning mechanism responsible for the replenishment of intracellular stores. Hippocampal SOCE is regulated by store-operated channels (SOC) organized in tripartite complex TRPC6/ORAI2/STIM2. It is suggested that in neurons, SOCE maintains intracellular homeostatic Ca2+ concentration at resting conditions and is needed to support the structure of dendritic spines. Recent evidence suggests that positive modulators of SOC are prospective drug candidates to treat Alzheimer's disease (AD) at early stages. Although STIM2 and ORAI2 are definitely involved in the regulation of nSOC amplitude and a play major role in AD pathogenesis, growing evidence suggest that it is not easy to target these proteins pharmacologically. Existing positive modulators of TRPC6 are unsuitable for drug development due to either bad pharmacokinetics or side effects. Thus, we concentrate the review on perspectives to develop specific nSOC modulators based on available 3D structures of TRPC6, ORAI2, and STIM2. We shortly describe the structural features of existing models and the methods used to prepare them. We provide commonly used steps applied for drug design based on 3D structures of target proteins that might be used to develop novel AD preventing therapy.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Diseño de Fármacos , Proteína ORAI2/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Canal Catiónico TRPC6/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Descubrimiento de Drogas , Humanos , Proteína ORAI2/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Molécula de Interacción Estromal 2/química , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Canal Catiónico TRPC6/química
6.
Cells ; 10(11)2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34831241

RESUMEN

Store-operated calcium entry (SOCE) provided through channels formed by ORAI proteins is a major regulator of several cellular processes. In immune cells, it controls fundamental processes such as proliferation, cell adhesion, and migration, while in cancer, SOCE and ORAI1 gene expression are dysregulated and lead to abnormal migration and/or cell proliferation. In the present study, we used the CRISPR/Cas9 technique to delete the ORAI1 gene and to identify its role in proliferative and migrative properties of the model cell line HEK-293. We showed that ORAI1 deletion greatly reduced SOCE. Thereby, we found that this decrease and the absence of ORAI1 protein did not affect HEK-293 proliferation. In addition, we determined that ORAI1 suppression did not affect adhesive properties but had a limited impact on HEK-293 migration. Overall, we showed that ORAI1 and SOCE are largely dispensable for cellular proliferation, migration, and cellular adhesion of HEK-293 cells. Thus, despite its importance in providing Ca2+ entry in non-excitable cells, our results indicate that the lack of SOCE does not deeply impact HEK-293 cells. This finding suggests the existence of compensatory mechanism enabling the maintenance of their physiological function.


Asunto(s)
Calcio/metabolismo , Movimiento Celular , Técnicas de Inactivación de Genes , Proteína ORAI1/deficiencia , Canales de Calcio/genética , Canales de Calcio/metabolismo , Adhesión Celular , Proliferación Celular , Genoma Humano , Células HEK293 , Humanos , Proteína ORAI1/metabolismo , Proteína ORAI2/genética , Proteína ORAI2/metabolismo
7.
Cell Rep ; 34(9): 108760, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657364

RESUMEN

Stromal-interaction molecules (STIM1/2) sense endoplasmic reticulum (ER) Ca2+ depletion and activate Orai channels. However, the choreography of interactions between native STIM/Orai proteins under physiological agonist stimulation is unknown. We show that the five STIM1/2 and Orai1/2/3 proteins are non-redundant and function together to ensure the graded diversity of mammalian Ca2+ signaling. Physiological Ca2+ signaling requires functional interactions between STIM1/2, Orai1/2/3, and IP3Rs, ensuring that receptor-mediated Ca2+ release is tailored to Ca2+ entry and nuclear factor of activated T cells (NFAT) activation. The N-terminal Ca2+-binding ER-luminal domains of unactivated STIM1/2 inhibit IP3R-evoked Ca2+ release. A gradual increase in agonist intensity and STIM1/2 activation relieves IP3R inhibition. Concomitantly, activated STIM1/2 C termini differentially interact with Orai1/2/3 as agonist intensity increases. Thus, coordinated and omnitemporal functions of all five STIM/Orai and IP3Rs translate the strength of agonist stimulation to precise levels of Ca2+ signaling and NFAT induction, ensuring the fidelity of complex mammalian Ca2+ signaling.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Proteína ORAI2/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Canales de Calcio/genética , Señalización del Calcio/efectos de los fármacos , Carbacol/farmacología , Células HEK293 , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Potenciales de la Membrana , Modelos Biológicos , Agonistas Muscarínicos/farmacología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Proteína ORAI1/genética , Proteína ORAI2/genética , Unión Proteica , Receptor Cross-Talk , Molécula de Interacción Estromal 1/agonistas , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 2/agonistas , Molécula de Interacción Estromal 2/genética , Factores de Tiempo
8.
Cell Calcium ; 91: 102281, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32896813

RESUMEN

The ubiquitous Ca2+ release-activated Ca2+ (CRAC) channel is crucial to many physiological functions. Both gain and loss of CRAC function is linked to disease. While ORAI1 is a crucial subunit of CRAC channels, recent evidence suggests that ORAI2 and ORAI3 heteromerize with ORAI1 to form native CRAC channels. Furthermore, ORAI2 and ORAI3 can form CRAC channels independently of ORAI1, suggesting diverse native CRAC stoichiometries. Yet, most available CRAC modifiers are presumed to target ORAI1 with little knowledge of their effects on ORAI2/3 or heteromers of ORAIs. Here, we used ORAI1/2/3 triple-null cells to express individual ORAI1, ORAI2, ORAI3 or ORAI1/2/3 concatemers. We reveal that GSK-7975A and BTP2 essentially abrogate ORAI1 and ORAI2 activity while causing only a partial inhibition of ORAI3. Interestingly, Synta66 abrogated ORAI1 channel function, while potentiating ORAI2 with no effect on ORAI3. CRAC channel activities mediated by concatenated ORAI1-1, ORAI1-2 and ORAI1-3 dimers were inhibited by Synta66, while ORAI2-3 dimers were unaffected. The CRAC enhancer IA65 significantly potentiated ORAI1 and ORAI1-1 activity with marginal effects on other ORAIs. Further, we characterized the profiles of individual ORAI isoforms in the presence of Gd3+ (5µM), 2-APB (5 µM and 50 µM), as well as changes in intracellular and extracellular pH. Our data reveal unique pharmacological features of ORAI isoforms expressed in an ORAI-null background and provide new insights into ORAI isoform selectivity of widely used CRAC pharmacological compounds.


Asunto(s)
Canales de Calcio/metabolismo , Proteína ORAI1/metabolismo , Proteína ORAI2/metabolismo , Anilidas/farmacología , Benzamidas/farmacología , Células HEK293 , Humanos , Isoformas de Proteínas/metabolismo , Multimerización de Proteína/efectos de los fármacos , Pirazoles/farmacología , Tiadiazoles/farmacología
9.
Life Sci ; 261: 118372, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32882268

RESUMEN

Despite remarkable progress in understanding and treating oral cancer (OC), it still remains one of the life-threatening diseases and predominant cancers in the world. Therefore, deciphering the molecular mechanisms of this disease would help us to develop highly efficacious therapies. Multiple lines of evidence suggest that calcium and its dysregulation play significant role in the development of various cancers. As an adaptation of survival mechanism, upon depletion of ER calcium stores, store-operated calcium entry (SOCE) has been induced via SOCE channels (SOCC) in various mammalian cells. SOCC are regulated by Orai-1, Orai-2 and Orai-3 located on plasma membrane and two calcium-sensing ER membrane proteins known as stromal interaction molecules (STIM-1 and STIM-2). Hence, the present study was aimed at analysing the role of Orai-1 and Orai-2 in oral cancer and the underlying mechanism. Our results suggest that both Orai-1 and Orai-2 proteins were overexpressed in oral cancer tissues and cell lines (SAS) compared to normal epithelial tissues and cell lines respectively. In addition, silencing of Orai-1 and Orai-2 via chemical SOCE inhibitors and siRNAs inhibited calcium uptake and suppressed oral cancer cell proliferation, colony formation and migration. Furthermore, silencing of Orai-1 and Orai-2 inhibited Akt/mTOR/NF-κB pathway in oral cancer cells. Interestingly, tobacco carcinogen NNN and synthetic carcinogen 4-NQO, enhanced the expression of Orai-1 and Orai-2 in SAS cells. Therefore, we conclude that Orai-1 and Orai-2 have significant role in oral cancer and can be further explored to develop novel therapies for the treatment of this disease.


Asunto(s)
Movimiento Celular , Neoplasias de la Boca/patología , FN-kappa B/metabolismo , Proteína ORAI1/metabolismo , Proteína ORAI2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Calcio/metabolismo , Señalización del Calcio , Carcinógenos/toxicidad , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Neoplasias de la Boca/genética , Transducción de Señal/efectos de los fármacos , Nicotiana/química , Ensayo de Tumor de Célula Madre
10.
Int J Mol Sci ; 21(15)2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32722509

RESUMEN

Senile plaques, the hallmarks of Alzheimer's Disease (AD), are generated by the deposition of amyloid-beta (Aß), the proteolytic product of amyloid precursor protein (APP), by ß and γ-secretase. A large body of evidence points towards a role for Ca2+ imbalances in the pathophysiology of both sporadic and familial forms of AD (FAD). A reduction in store-operated Ca2+ entry (SOCE) is shared by numerous FAD-linked mutations, and SOCE is involved in Aß accumulation in different model cells. In neurons, both the role and components of SOCE remain quite obscure, whereas in astrocytes, SOCE controls their Ca2+-based excitability and communication to neurons. Glial cells are also directly involved in Aß production and clearance. Here, we focus on the role of ORAI2, a key SOCE component, in modulating SOCE in the human neuroglioma cell line H4. We show that ORAI2 overexpression reduces both SOCE level and stores Ca2+ content, while ORAI2 downregulation significantly increases SOCE amplitude without affecting store Ca2+ handling. In Aß-secreting H4-APPswe cells, SOCE inhibition by BTP2 and SOCE augmentation by ORAI2 downregulation respectively increases and decreases Aß42 accumulation. Based on these findings, we suggest ORAI2 downregulation as a potential tool to rescue defective SOCE in AD, while preventing plaque formation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Señalización del Calcio , Neuronas/metabolismo , Proteína ORAI2/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/patología , Células HEK293 , Células HeLa , Humanos , Neuronas/patología
11.
Biochim Biophys Acta Mol Cell Res ; 1867(1): 118567, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676354

RESUMEN

Acinar cell exocytosis requires spatiotemporal Ca2+ signals regulated through endoplasmic reticulum (ER) stores, Ca2+ATPases, and store-operated Ca2+ entry (SOCE). The secretory pathway Ca2+ATPase 2 (SPCA2) interacts with Orai1, which is involved in SOCE and store independent Ca2+ entry (SICE). However, in the pancreas, only a C-terminally truncated form of SPCA2 (termed SPAC2C) exists. The goal of this study was to determine if SPCA2C effects Ca2+ homeostasis in a similar fashion to the full-length SPCA2. Using epitope-tagged SPCA2C (SPCA2CFLAG) expressed in HEK293A cells and Fura2 imaging, cytosolic [Ca2+] was examined during SICE, SOCE and secretagogue-stimulated signaling. Exogenous SPCA2C expression increased resting cytosolic [Ca2+], Ca2+ release in response to carbachol, ER Ca2+ stores, and store-mediated and independent Ca2+ influx. Co-IP detected Orai1-SPCA2C interaction, which was altered by co-expression of STIM1. Importantly, SPCA2C's effects on store-mediated Ca2+ entry were independent of Orai1. These findings indicate SPCA2C influences Ca2+ homeostasis through multiple mechanisms, some of which are independent of Orai1, suggesting novel and possibly cell-specific Ca2+ regulation.


Asunto(s)
Señalización del Calcio/fisiología , ATPasas Transportadoras de Calcio/fisiología , Calcio/metabolismo , Páncreas/metabolismo , Canales de Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Células HEK293 , Homeostasis , Humanos , Proteína ORAI2/genética , Proteína ORAI2/metabolismo , Especificidad de Órganos/genética , Isoformas de Proteínas/fisiología , Vías Secretoras/fisiología
12.
Dis Markers ; 2019: 7049294, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31772693

RESUMEN

BACKGROUND: Ca2+ release-activated Ca2+ channels (CRAC) are the main Ca2+ entry pathway regulating intracellular Ca2+ concentration in a variety of cancer types. Orai2 is the main pore-forming subunit of CRAC channels in central neurons. To explore the role of Orai2 in glioblastoma (GBM), we investigated the key pathways and genes in Orai2-mediated GBM by bioinformatic analyses. METHODS: Via The Cancer Genome Atlas (TCGA), French, Sun, and Gene Expression Omnibus (GEO) (GDS3885) datasets, we collected 1231 cases with RNA-seq data and analyzed the functional annotation of Orai2 by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Univariate and multivariate survival analyses were applied to 823 patients with survival data. RESULTS: We discovered that Orai2 was markedly upregulated in GBM compared to normal brain samples and lower-grade gliomas (LGG). Survival analysis found that higher expression of Orai2 was independently associated with a worse prognosis of patients with the classical and mesenchymal subtypes of GBM. Simultaneously, Orai2 expression was higher in tumors of the classical and mesenchymal subtypes than other subtypes and was significantly correlated with classical- and mesenchymal-related genes. GO and KEGG pathway analysis revealed that genes significantly correlated with Orai2 were involved in the JNK pathway. Through screening transcriptomic data, we found a strong association between Orai2 and apoptosis, stemness, and an epithelial-mesenchymal transition- (EMT-) like phenotype. CONCLUSION: As a prognostic factor, Orai2 is obviously activated in the classical and mesenchymal subtypes of GBM and promotes glioma cell self-renewal, apoptosis, and EMT-like by the JNK pathway. These findings indicate that Orai2 could be a candidate prognostic and therapeutic target, especially for the classical and mesenchymal subtypes of GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Proteína ORAI2/genética , Apoptosis/genética , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Análisis por Conglomerados , Bases de Datos Factuales , Transición Epitelial-Mesenquimal/genética , Glioblastoma/genética , Glioblastoma/mortalidad , Humanos , Estimación de Kaplan-Meier , Sistema de Señalización de MAP Quinasas/genética , Proteína ORAI2/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico
13.
Stroke ; 50(11): 3238-3245, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31551038

RESUMEN

Background and Purpose- Ischemic stroke is one of the leading causes of disability and death. The principal goal of acute stroke treatment is the recanalization of the occluded cerebral arteries, which is, however, only effective in a very narrow time window. Therefore, neuroprotective treatments that can be combined with recanalization strategies are needed. Calcium overload is one of the major triggers of neuronal cell death. We have previously shown that capacitative Ca2+ entry, which is triggered by the depletion of intracellular calcium stores, contributes to ischemia-induced calcium influx in neurons, but the responsible Ca2+ channel is not known. Methods- Here, we have generated mice lacking the calcium channel subunit Orai2 and analyzed them in experimental stroke. Results- Orai2-deficient mice were protected from ischemic neuronal death both during acute ischemia under vessel occlusion and during ischemia/reperfusion upon successful recanalization. Calcium signals induced by calcium store depletion or oxygen/glucose deprivation were significantly diminished in Orai2-deficient neurons demonstrating that Orai2 is a central mediator of neuronal capacitative Ca2+ entry and is involved in calcium overload during ischemia. Conclusions- Our experimental data identify Orai2 as an attractive target for pharmaceutical intervention in acute stroke.


Asunto(s)
Isquemia Encefálica , Señalización del Calcio , Calcio/metabolismo , Neuroprotección , Proteína ORAI2/deficiencia , Accidente Cerebrovascular , Enfermedad Aguda , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Muerte Celular , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Proteína ORAI2/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/prevención & control
14.
Nat Commun ; 10(1): 3223, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31324793

RESUMEN

It is widely assumed that inositol trisphosphate (IP3) and ryanodine (Ry) receptors share the same Ca2+ pool in central mammalian neurons. We now demonstrate that in hippocampal CA1 pyramidal neurons IP3- and Ry-receptors are associated with two functionally distinct intracellular Ca2+ stores, respectively. While the IP3-sensitive Ca2+ store refilling requires Orai2 channels, Ry-sensitive Ca2+ store refilling involves voltage-gated Ca2+ channels (VGCCs). Our findings have direct implications for the understanding of function and plasticity in these central mammalian neurons.


Asunto(s)
Calcio/metabolismo , Hipocampo/metabolismo , Proteína ORAI2/metabolismo , Células Piramidales/metabolismo , Animales , Canales de Calcio , Regulación de la Expresión Génica , Fosfatos de Inositol/metabolismo , Iones , Ratones , Ratones Noqueados , Modelos Animales , Proteína ORAI2/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
15.
Br J Pharmacol ; 176(19): 3845-3856, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271653

RESUMEN

BACKGROUND AND PURPOSE: Mibefradil, a T-type Ca2+ channel blocker, has been investigated for treating solid tumours. However, its underlying mechanisms are still unclear. Here, we have investigated the pharmacological actions of mibefradil on Orai store-operated Ca2+ channels. EXPERIMENTAL APPROACH: Human Orai1-3 cDNAs in tetracycline-regulated pcDNA4/TO vectors were transfected into HEK293 T-REx cells with stromal interaction molecule 1 (STIM1) stable expression. The Orai currents were recorded by whole-cell and excised-membrane patch clamp. Ca2+ influx or release was measured by Fura-PE3/AM. Cell growth and death were monitored by WST-1, LDH assays and flow cytometry. KEY RESULTS: Mibefradil inhibited Orai1, Orai2, and Orai3 currents dose-dependently. The IC50 for Orai1, Orai2, and Orai3 channels was 52.6, 14.1, and 3.8 µM respectively. Outside-out patch demonstrated that perfusion of 10-µM mibefradil to the extracellular surface completely blocked Orai3 currents and single channel activity evoked by 2-APB. Intracellular application of mibefradil did not alter Orai3 channel activity. Mibefradil at higher concentrations (>50 µM) inhibited Ca2+ release but had no effect on cytosolic STIM1 translocation evoked by thapsigargin. Inhibition on Orai channels by mibefradil was structure-related, as other T-type Ca2+ channel blockers with different structures, such as ethosuximide and ML218, had no or minimal effects on Orai channels. Moreover, mibefradil inhibited cell proliferation, induced apoptosis, and arrested cell cycle progression. CONCLUSIONS AND IMPLICATIONS: Mibefradil is a potent cell surface blocker of Orai channels, demonstrating a new pharmacological action of this compound in regulating cell growth and death, which could be relevant to its anti-cancer activity.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Mibefradil/farmacología , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI2/antagonistas & inhibidores , Calcio/análisis , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/química , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células HEK293 , Humanos , Mibefradil/química , Proteína ORAI1/metabolismo , Proteína ORAI2/metabolismo , Imagen Óptica
16.
FASEB J ; 33(9): 9775-9784, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31166814

RESUMEN

The leucine-rich repeat kinase 2 (LRRK2) is expressed in various immune cells and involved in regulating inflammatory processes. LRRK2 facilitates calcium extrusion exchanger and sodium-calcium exchanger activity and hence influences intracellular Ca2+ concentration in dendritic cells (DCs). DC maturation and migration are governed by the intracellular Ca2+ concentration, but the related mechanisms whereby LRRK2 regulates DC function and involved Ca2+ channels are still under investigation. In the previous study, we found that LRRK2-/- DCs exhibited higher store-operated Ca2+ entry (SOCE) activity than LRRK2+/+ DCs. Herein, we ascertained the exact SOCE components by using genetic, pharmacological, and fluorescent approaches. Ca2+ imaging showed that LRRK2 kinase activity negatively modulated SOCE activity. Moreover, LRRK2 deficiency resulted in an enhanced migration capacity of DCs but had little effect on the maturation process. SOCE is widely known to regulate DC functions; we wanted to dissect the reason why LRRK2 specifically influenced DC migration and therefore silenced ORAI1, ORAI2, and ORAI3, respectively. Transwell assays showed that both ORAI1 and ORAI2 silencing markedly decreased the migration of DCs, but only ORAI1 deficiency influenced the expression of maturation markers CD11c, CD86, and major histocompatibility complex class II. Of note, LRRK2 deficiency increased ORAI2 expression but not that of ORAI1 and ORAI3. Thus, we suggest that LRRK2 modulates DC migration by interfering with ORAI2.-Yan, J., Zhao, W., Gao, C., Liu, X., Zhao, X., Wei, T., Gao, Z. Leucine-rich repeat kinase 2 regulates mouse dendritic cell migration by ORAI2.


Asunto(s)
Movimiento Celular/fisiología , Células Dendríticas/fisiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteína ORAI2/metabolismo , Animales , Compuestos de Boro/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Silenciador del Gen , Imidazoles/farmacología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Proteína ORAI2/genética , Molécula de Interacción Estromal 2/genética , Molécula de Interacción Estromal 2/metabolismo
17.
Biochim Biophys Acta Mol Cell Res ; 1866(7): 1137-1150, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30659848

RESUMEN

Orai proteins form highly selective Ca2+ release-activated channels (CRACs). They play a critical role in store-operated Ca2+ entry (SOCE; i.e., the influx of external Ca2+ that is induced by the depletion of endoplasmic reticulum Ca2+ stores). Of the three Orai homologs that are present in mammals (Orai1-3), the physiological function of Orai1 is the best described. CRACs are formed by both homomeric assemblies and heteromultimers of Orais. Orai1 and Orai2 can form heteromeric channels that differ in conductivity during SOCE, depending on their Orai1-to-Orai2 ratio. The present study explored the potential consequences of ORAI1 overexpression in neurons where the dominant isoform is Orai2. We established the Tg(ORAI1)Ibd transgenic mouse line that overexpresses ORAI1 in brain neurons. We observed seizure-like symptoms in aged (≥15-month-old) female mice but not in males of the same age. The application of kainic acid and bicuculline to slices that were isolated from 8-month-old (±1 month) female Tg(ORAI1)Ibd mice revealed a significantly lower frequency of interictal bursts compared with samples that were isolated from wildtype mice. No differences were observed in male mice of a similar age. A battery of behavioral tests showed that context recognition decreased only in female transgenic mice. The phenotype that was observed in female mice suggests that ORAI1 overexpression may affect neuronal activity in a sex-dependent manner. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.


Asunto(s)
Conducta Animal , Ondas Encefálicas , Encéfalo/metabolismo , Neuronas/metabolismo , Proteína ORAI1/biosíntesis , Proteína ORAI2/metabolismo , Convulsiones/metabolismo , Animales , Encéfalo/patología , Femenino , Ratones , Ratones Transgénicos , Neuronas/patología , Proteína ORAI1/genética , Proteína ORAI2/genética , Convulsiones/genética , Convulsiones/patología , Convulsiones/fisiopatología
18.
Nat Commun ; 8(1): 1920, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29203863

RESUMEN

Impaired albumin reabsorption by proximal tubular epithelial cells (PTECs) has been highlighted in diabetic nephropathy (DN), but little is known about the underlying molecular mechanisms. Here we find that ORAI1-3, are preferentially expressed in PTECs and downregulated in patients with DN. Hyperglycemia or blockade of insulin signaling reduces the expression of ORAI1-3. Inhibition of ORAI channels by BTP2 and diethylstilbestrol or silencing of ORAI expression impairs albumin uptake. Transgenic mice expressing a dominant-negative Orai1 mutant (E108Q) increases albuminuria, and in vivo injection of BTP2 exacerbates albuminuria in streptozotocin-induced and Akita diabetic mice. The albumin endocytosis is Ca2+-dependent and accompanied by ORAI1 internalization. Amnionless (AMN) associates with ORAIs and forms STIM/ORAI/AMN complexes after Ca2+ store depletion. STIM1/ORAI1 colocalizes with clathrin, but not with caveolin, at the apical membrane of PTECs, which determines clathrin-mediated endocytosis. These findings provide insights into the mechanisms of protein reabsorption and potential targets for treating diabetic proteinuria.


Asunto(s)
Albúminas/metabolismo , Albuminuria/genética , Canales de Calcio/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatías Diabéticas/metabolismo , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Proteína ORAI1/genética , Proteína ORAI2/genética , Albúminas/efectos de los fármacos , Albuminuria/metabolismo , Anilidas/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio , Canales de Calcio/metabolismo , Estudios de Casos y Controles , Caveolinas/metabolismo , Línea Celular , Clatrina/metabolismo , Dietilestilbestrol/farmacología , Regulación hacia Abajo , Endocitosis , Células Epiteliales/efectos de los fármacos , Estrógenos no Esteroides/farmacología , Femenino , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Riñón/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Mutación , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI1/metabolismo , Proteína ORAI2/antagonistas & inhibidores , Proteína ORAI2/metabolismo , Reabsorción Renal/efectos de los fármacos , Reabsorción Renal/genética , Molécula de Interacción Estromal 1/metabolismo , Tiadiazoles/farmacología
19.
Cell Calcium ; 66: 33-47, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28807148

RESUMEN

Basal forebrain neurons increase cortical blood flow by releasing acetylcholine (Ach), which stimulates endothelial cells (ECs) to produce the vasodilating gasotransmitter, nitric oxide (NO). Surprisingly, the mechanism whereby Ach induces NO synthesis in brain microvascular ECs is unknown. An increase in intracellular Ca2+ concentration recruits a multitude of endothelial Ca2+-dependent pathways, such as Ca2+/calmodulin endothelial NO synthase (eNOS). The present investigation sought to investigate the role of intracellular Ca2+ signaling in Ach-induced NO production in bEND5 cells, an established model of mouse brain microvascular ECs, by conventional imaging of cells loaded with the Ca2+-sensitive dye, Fura-2/AM, and the NO-sensitive fluorophore, DAF-DM diacetate. Ach induced dose-dependent Ca2+ oscillations in bEND5 cells, 300 µM being the most effective dose to generate a prolonged Ca2+ burst. Pharmacological manipulation revealed that Ach-evoked Ca2+ oscillations required metabotropic muscarinic receptor (mAchR) activation and were patterned by a complex interplay between repetitive ER Ca2+ release via inositol-1,4,5-trisphosphate receptors (InsP3Rs) and store-operated Ca2+ entry (SOCE). A comprehensive real time-polymerase chain reaction analysis demonstrated the expression of the transcripts encoding for M3-mAChRs, InsP3R1 and InsP3R3, Stim1-2 and Orai2. Next, we found that Ach-induced NO production was hindered by L-NAME, a selective NOS inhibitor, and BAPTA, a membrane permeable intracellular Ca2+ buffer. Moreover, Ach-elicited NO synthesis was blocked by the pharmacological abrogation of the accompanying Ca2+ spikes. Overall, these data shed novel light on the molecular mechanisms whereby neuronally-released Ach controls neurovascular coupling in blood microvessels.


Asunto(s)
Acetilcolina/farmacología , Señalización del Calcio/efectos de los fármacos , Óxido Nítrico/metabolismo , Animales , Encéfalo/citología , Calcio/metabolismo , Línea Celular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fura-2/química , Fura-2/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Microvasos/citología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína ORAI2/genética , Proteína ORAI2/metabolismo , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo
20.
FASEB J ; 31(8): 3439-3448, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28446591

RESUMEN

The transcription factor nuclear factor of activated T cells 5 (NFAT5) is up-regulated in several clinical disorders, including dehydration. NFAT5-sensitive genes include serum and glucocorticoid-inducible kinase 1 (SGK1). The kinase is a powerful regulator of Orai1, a Ca2+ channel accomplishing store-operated Ca2+ entry (SOCE). Orai1 is stimulated after intracellular store depletion by the Ca2+ sensors stromal interaction molecule 1 (STIM1), or STIM2, or both. In the present study, we explored whether nuclear factor of activated T cell (NFAT)-5 influences Ca2+ signaling in megakaryocytes. To this end, human megakaryocytic (MEG-01) cells were transfected with NFAT5 or with siNFAT5. Platelets and megakaryocytes were isolated from wild-type mice with either access to water ad libitum or dehydration by 36 h of water deprivation. Transcript levels were determined with quantitative RT-PCR and protein abundance by Western blot analysis and flow cytometry, cytosolic (intracellular) Ca2+ concentration ([Ca2+]i) by fura-2-fluorescence. SOCE was estimated from the increase of [Ca2+]i following readdition of extracellular Ca2+ after store depletion with thapsigargin (1 µM). Platelet degranulation was estimated from P-selectin abundance and integrin activation from αIIbß3 integrin abundance determined by flow cytometry. As a result, NFAT5 transfection or exposure to hypertonicity (+40 mM NaCl) of MEG-01 cells increased Orai1, Orai2, STIM1, and STIM2 transcript levels. Orai1 transcript levels were decreased by NFAT5 silencing. NFAT5 transfection and IκB inhibitor BMS 345541 (5 µM) increased SOCE, whereas NFAT5 silencing and SGK1 inhibitor GSK650394 (10 µM) decreased SOCE. In the mice, dehydration increased NFAT5 and Orai1 protein abundance in megakaryocytes and NFAT5, Orai1, and Orai2 abundance in platelets. Dehydration further augmented the degranulation and integrin activation by thrombin and collagen-related peptide. In summary, NFAT5 is a powerful regulator of Orai1-expression and SOCE in megakaryocytes.-Sahu, I., Pelzl, L., Sukkar, B., Fakhri, H., al-Maghout, T., Cao, H., Hauser, S., Gutti, R., Gawaz, M., Lang, F. NFAT5-sensitive Orai1 expression and store-operated Ca2+ entry in megakaryocytes.


Asunto(s)
Calcio/metabolismo , Megacariocitos/metabolismo , Proteína ORAI1/metabolismo , Proteína ORAI2/metabolismo , Factores de Transcripción/metabolismo , Animales , Plaquetas , Línea Celular , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Proteína ORAI1/genética , Proteína ORAI2/genética , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/genética , Molécula de Interacción Estromal 2/metabolismo , Factores de Transcripción/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...