Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Oncogene ; 42(24): 1980-1993, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37130916

ABSTRACT

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.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Methyltransferases/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/secondary , RNA Processing, Post-Transcriptional , ORAI2 Protein/genetics , ORAI2 Protein/metabolism
2.
Metab Brain Dis ; 38(4): 1297-1310, 2023 04.
Article in English | MEDLINE | ID: mdl-36809524

ABSTRACT

The progressive, chronic nature of Alzheimer's disease (AD), a form of dementia, defaces the adulthood of elderly individuals. The pathogenesis of the condition is primarily unascertained, turning the treatment efficacy more arduous. Therefore, understanding the genetic etiology of AD is essential to identifying targeted therapeutics. This study aimed to use machine-learning techniques of expressed genes in patients with AD to identify potential biomarkers that can be used for future therapy. The dataset is accessed from the Gene Expression Omnibus (GEO) database (Accession Number: GSE36980). The subgroups (AD blood samples from frontal, hippocampal, and temporal regions) are individually investigated against non-AD models. Prioritized gene cluster analyses are conducted with the STRING database. The candidate gene biomarkers were trained with various supervised machine-learning (ML) classification algorithms. The interpretation of the model prediction is perpetrated with explainable artificial intelligence (AI) techniques. This experiment revealed 34, 60, and 28 genes as target biomarkers of AD mapped from the frontal, hippocampal, and temporal regions. It is identified ORAI2 as a shared biomarker in all three areas strongly associated with AD's progression. The pathway analysis showed that STIM1 and TRPC3 are strongly associated with ORAI2. We found three hub genes, TPI1, STIM1, and TRPC3, in the network of the ORAI2 gene that might be involved in the molecular pathogenesis of AD. Naive Bayes classified the samples of different groups by fivefold cross-validation with 100% accuracy. AI and ML are promising tools in identifying disease-associated genes that will advance the field of targeted therapeutics against genetic diseases.


Subject(s)
Alzheimer Disease , Humans , Adult , Aged , Alzheimer Disease/metabolism , Artificial Intelligence , Bayes Theorem , Computational Biology/methods , Biomarkers , Gene Expression , ORAI2 Protein/genetics
3.
Genomics ; 114(5): 110479, 2022 09.
Article in English | MEDLINE | ID: mdl-36070824

ABSTRACT

Orai2 is a component of store-operated Calcium channels (SOCCs) and exerts a pivotal role in immunity. In intestinal macrophages (Mφs), Orai2 deficiency influenced linoleic acid (LA)-arachidonic acid (ARA) derivatives by regulating Pla2g6 and Alox5. 16S rRNA sequencing showed that deleting Orai2 facilitated the prevalence of Akkermansia muciniphila, and untargeted metabolomics confirmed the suppressed level of leukotriene A. Moreover, Orai2 deficiency ameliorated the progression of experimental murine colitis, as shown by attenuated structural collapse of colon and pro-inflammatory cytokine concentrations, and rescued dysbiosis. The administration of a Pla2g6 inhibitor (Bromoenol lactone) not only inhibited the relative abundance of A. muciniphila in the feces of Orai2 knockout (Orai2-/-) mice, but also abolished the increased activity of Calcium-released activated Calcium channel (CRAC) in Orai2-/- intestinal Mφs, corroborating the involvement of Pla2g6 in Orai2 signaling. In conclusion, Orai2 deficiency increases Pla2g6 and hence facilitating A. muciniphila colonization, which might be a potential strategy to combat colitis.


Subject(s)
Calcium , Colitis , Akkermansia , Animals , Arachidonic Acid , Calcium/metabolism , Calcium Channels/genetics , Colitis/genetics , Cytokines , Group VI Phospholipases A2 , Leukotriene A4 , Linoleic Acid , Mice , ORAI2 Protein/genetics , RNA, Ribosomal, 16S
4.
Int J Mol Sci ; 23(16)2022 Aug 22.
Article in English | MEDLINE | ID: mdl-36012752

ABSTRACT

During ischemic stroke, infarct growth before recanalization diminishes functional outcome. Hence, adjunct treatment options to protect the ischemic penumbra before recanalization are eagerly awaited. In experimental stroke targeting two different pathways conferred protection from penumbral tissue loss: (1) enhancement of hypoxic tolerance of neurons by deletion of the calcium channel subunit Orai2 and (2) blocking of detrimental lymphocyte-platelet responses. However, until now, no preclinical stroke study has assessed the potential of combining neuroprotective with anti-thrombo-inflammatory interventions to augment therapeutic effects. We induced focal cerebral ischemia in Orai2-deficient (Orai2-/-) mice by middle cerebral artery occlusion (MCAO). Animals were treated with anti-glycoprotein Ib alpha (GPIbα) Fab fragments (p0p/B Fab) blocking GPIbα-von Willebrand factor (vWF) interactions. Rat immunoglobulin G (IgG) Fab was used as the control treatment. The extent of infarct growth before recanalization was assessed at 4 h after MCAO. Moreover, infarct volumes were determined 6 h after recanalization (occlusion time: 4 h). Orai2 deficiency significantly halted cerebral infarct progression under occlusion. Inhibition of platelet GPIbα further reduced primary infarct growth in Orai2-/- mice. During ischemia-reperfusion, upon recanalization, mice were likewise protected. All in all, we show that neuroprotection in Orai2-/- mice can be augmented by targeting thrombo-inflammation. This supports the clinical development of combined neuroprotective/anti-platelet strategies in hyper-acute stroke.


Subject(s)
Brain Ischemia , ORAI2 Protein , Stroke , Animals , Blood Platelets/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Brain Ischemia/prevention & control , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Mice , Mice, Knockout , Neuroprotection , ORAI2 Protein/genetics , Stroke/metabolism
5.
Glia ; 70(9): 1666-1680, 2022 09.
Article in English | MEDLINE | ID: mdl-35506586

ABSTRACT

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.


Subject(s)
Astrocytes , Interleukin-1alpha , ORAI2 Protein , Prostaglandins E , Tumor Necrosis Factor-alpha , Animals , Astrocytes/metabolism , Calcium/metabolism , Calcium Signaling , Inflammation , Interleukin-1alpha/metabolism , Interleukin-1alpha/pharmacology , Mice , ORAI2 Protein/metabolism , Prostaglandins E/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
J Physiol ; 600(3): 623-643, 2022 02.
Article in English | MEDLINE | ID: mdl-34877682

ABSTRACT

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 .


Subject(s)
Calcium Channels , Calcium Release Activated Calcium Channels , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , HEK293 Cells , Humans , Hydrogen-Ion Concentration , ORAI1 Protein/genetics , ORAI2 Protein/metabolism , Stromal Interaction Molecule 1/metabolism
7.
Nat Commun ; 12(1): 7035, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857756

ABSTRACT

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.


Subject(s)
Alzheimer Disease/genetics , ORAI2 Protein/genetics , RNA Editing , RNA/genetics , Synaptotagmins/genetics , Transcriptome , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Atlases as Topic , Brain/metabolism , Brain/pathology , Brain Chemistry , Gene Expression Profiling , Humans , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , ORAI2 Protein/metabolism , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism , RNA/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Synaptotagmins/metabolism
8.
Int J Mol Sci ; 22(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34948414

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Drug Design , ORAI2 Protein/metabolism , Stromal Interaction Molecule 2/metabolism , TRPC6 Cation Channel/metabolism , Alzheimer Disease/metabolism , Animals , Drug Discovery , Humans , ORAI2 Protein/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Stromal Interaction Molecule 2/chemistry , Synapses/drug effects , Synapses/metabolism , TRPC6 Cation Channel/chemistry
9.
Cells ; 10(11)2021 11 04.
Article in English | MEDLINE | ID: mdl-34831241

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Cell Movement , Gene Knockout Techniques , ORAI1 Protein/deficiency , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Adhesion , Cell Proliferation , Genome, Human , HEK293 Cells , Humans , ORAI1 Protein/metabolism , ORAI2 Protein/genetics , ORAI2 Protein/metabolism
10.
Cell Rep ; 34(9): 108760, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33657364

ABSTRACT

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.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , ORAI2 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/metabolism , Calcium Channels/genetics , Calcium Signaling/drug effects , Carbachol/pharmacology , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Potentials , Models, Biological , Muscarinic Agonists/pharmacology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , ORAI2 Protein/genetics , Protein Binding , Receptor Cross-Talk , Stromal Interaction Molecule 1/agonists , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 2/agonists , Stromal Interaction Molecule 2/genetics , Time Factors
11.
Cancer Res ; 81(4): 986-1000, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33310726

ABSTRACT

The ubiquitous second messenger Ca2+ has long been recognized as a key regulator in cell migration. Locally confined Ca2+, in particular, is essential for building front-to-rear Ca2+ gradient, which serves to maintain the morphologic polarity required in directionally migrating cells. However, little is known about the source of the Ca2+ and the mechanism by which they crosstalk between different signaling pathways in cancer cells. Here, we report that calcium release-activated calcium modulator 2 (ORAI2), a poorly characterized store-operated calcium (SOC) channel subunit, predominantly upregulated in the lymph node metastasis of gastric cancer, supports cell proliferation and migration. Clinical data reveal that a high frequency of ORAI2-positive cells in gastric cancer tissues significantly correlated with poor differentiation, invasion, lymph node metastasis, and worse prognosis. Gain- and loss-of-function showed that ORAI2 promotes cell motility, tumor formation, and metastasis in both gastric cancer cell lines and mice. Mechanistically, ORAI2 mediated SOC activity and regulated tumorigenic properties through the activation of the PI3K/Akt signaling pathways. Moreover, ORAI2 enhanced the metastatic ability of gastric cancer cells by inducing FAK-mediated MAPK/ERK activation and promoted focal adhesion disassembly at rear-edge of the cell. Collectively, our results demonstrate that ORAI2 is a novel gene that plays an important role in the tumorigenicity and metastasis of gastric cancer. SIGNIFICANCE: These findings describe the critical role of ORAI2 in gastric cancer cell migration and tumor metastasis and uncover the translational potential to advance drug discovery along the ORAI2 signaling pathway.


Subject(s)
Adenocarcinoma/pathology , Carcinogenesis/genetics , Focal Adhesions/metabolism , ORAI2 Protein/physiology , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Focal Adhesions/genetics , Focal Adhesions/pathology , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , ORAI2 Protein/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
12.
Cell Calcium ; 91: 102281, 2020 11.
Article in English | MEDLINE | ID: mdl-32896813

ABSTRACT

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.


Subject(s)
Calcium Channels/metabolism , ORAI1 Protein/metabolism , ORAI2 Protein/metabolism , Anilides/pharmacology , Benzamides/pharmacology , HEK293 Cells , Humans , Protein Isoforms/metabolism , Protein Multimerization/drug effects , Pyrazoles/pharmacology , Thiadiazoles/pharmacology
13.
Life Sci ; 261: 118372, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32882268

ABSTRACT

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.


Subject(s)
Cell Movement , Mouth Neoplasms/pathology , NF-kappa B/metabolism , ORAI1 Protein/metabolism , ORAI2 Protein/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Calcium/metabolism , Calcium Signaling , Carcinogens/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Humans , Mouth Neoplasms/genetics , Signal Transduction/drug effects , Nicotiana/chemistry , Tumor Stem Cell Assay
14.
Proc Natl Acad Sci U S A ; 117(39): 24403-24414, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929002

ABSTRACT

Calcium signals are initiated in immune cells by the process of store-operated calcium entry (SOCE), where receptor activation triggers transient calcium release from the endoplasmic reticulum, followed by opening of plasma-membrane calcium-release activated calcium (CRAC) channels. ORAI1, ORAI2, and ORAI3 are known to comprise the CRAC channel; however, the contributions of individual isoforms to neutrophil function are not well understood. Here, we show that loss of ORAI1 partially decreases calcium influx, while loss of both ORAI1 and ORAI2 completely abolishes SOCE. In other immune-cell types, loss of ORAI2 enhances SOCE. In contrast, we find that ORAI2-deficient neutrophils display decreased calcium influx, which is correlated with measurable differences in the regulation of neutrophil membrane potential via KCa3.1. Decreased SOCE in ORAI1-, ORAI2-, and ORAI1/2-deficient neutrophils impairs multiple neutrophil functions, including phagocytosis, degranulation, leukotriene, and reactive oxygen species (ROS) production, rendering ORAI1/2-deficient mice highly susceptible to staphylococcal infection. This study demonstrates that ORAI1 and ORAI2 are the primary components of the neutrophil CRAC channel and identifies subpopulations of neutrophils where cell-membrane potential functions as a rheostat to modulate the SOCE response. These findings have implications for mechanisms that modulate neutrophil function during infection, acute and chronic inflammatory conditions, and cancer.


Subject(s)
Calcium Signaling , Calcium/immunology , Neutrophils/immunology , ORAI1 Protein/immunology , ORAI2 Protein/immunology , Animals , Female , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Male , Mice , Mice, Inbred C57BL , ORAI1 Protein/genetics , ORAI2 Protein/genetics
15.
Int J Mol Sci ; 21(15)2020 Jul 25.
Article in English | MEDLINE | ID: mdl-32722509

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Calcium Signaling , Neurons/metabolism , ORAI2 Protein/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/pathology , HEK293 Cells , HeLa Cells , Humans , Neurons/pathology
16.
Biochim Biophys Acta Mol Cell Res ; 1867(1): 118567, 2020 01.
Article in English | MEDLINE | ID: mdl-31676354

ABSTRACT

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.


Subject(s)
Calcium Signaling/physiology , Calcium-Transporting ATPases/physiology , Calcium/metabolism , Pancreas/metabolism , Calcium Channels/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Homeostasis , Humans , ORAI2 Protein/genetics , ORAI2 Protein/metabolism , Organ Specificity/genetics , Protein Isoforms/physiology , Secretory Pathway/physiology
17.
Dis Markers ; 2019: 7049294, 2019.
Article in English | MEDLINE | ID: mdl-31772693

ABSTRACT

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.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , ORAI2 Protein/genetics , Apoptosis/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cluster Analysis , Databases, Factual , Epithelial-Mesenchymal Transition/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , MAP Kinase Signaling System/genetics , ORAI2 Protein/metabolism , Oligonucleotide Array Sequence Analysis , Prognosis
18.
Stroke ; 50(11): 3238-3245, 2019 11.
Article in English | MEDLINE | ID: mdl-31551038

ABSTRACT

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.


Subject(s)
Brain Ischemia , Calcium Signaling , Calcium/metabolism , Neuroprotection , ORAI2 Protein/deficiency , Stroke , Acute Disease , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Cell Death , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , ORAI2 Protein/metabolism , Stroke/genetics , Stroke/metabolism , Stroke/pathology , Stroke/prevention & control
19.
Nat Commun ; 10(1): 3223, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324793

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , ORAI2 Protein/metabolism , Pyramidal Cells/metabolism , Animals , Calcium Channels , Gene Expression Regulation , Inositol Phosphates/metabolism , Ions , Mice , Mice, Knockout , Models, Animal , ORAI2 Protein/genetics , Ryanodine Receptor Calcium Release Channel/metabolism
20.
Br J Pharmacol ; 176(19): 3845-3856, 2019 10.
Article in English | MEDLINE | ID: mdl-31271653

ABSTRACT

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.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Mibefradil/pharmacology , ORAI1 Protein/antagonists & inhibitors , ORAI2 Protein/antagonists & inhibitors , Calcium/analysis , Calcium/metabolism , Calcium Channel Blockers/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Cells, Cultured , HEK293 Cells , Humans , Mibefradil/chemistry , ORAI1 Protein/metabolism , ORAI2 Protein/metabolism , Optical Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...