Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6921, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37903816

ABSTRACT

Ca2+ signal-generation through inter-membrane junctional coupling between endoplasmic reticulum (ER) STIM proteins and plasma membrane (PM) Orai channels, remains a vital but undefined mechanism. We identify two unusual overlapping Phe-His aromatic pairs within the STIM1 apical helix, one of which (F394-H398) mediates important control over Orai1-STIM1 coupling. In resting STIM1, this locus is deeply clamped within the folded STIM1-CC1 helices, likely near to the ER surface. The clamped environment in holo-STIM1 is critical-positive charge replacing Phe-394 constitutively unclamps STIM1, mimicking store-depletion, negative charge irreversibly locks the clamped-state. In store-activated, unclamped STIM1, Phe-394 mediates binding to the Orai1 channel, but His-398 is indispensable for transducing STIM1-binding into Orai1 channel-gating, and is spatially aligned with Phe-394 in the exposed Sα2 helical apex. Thus, the Phe-His locus traverses between ER and PM surfaces and is decisive in the two critical STIM1 functions-unclamping to activate STIM1, and conformational-coupling to gate the Orai1 channel.


Subject(s)
Calcium Signaling , Calcium , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Calcium Signaling/physiology
2.
Article in English | MEDLINE | ID: mdl-37317920

ABSTRACT

BACKGROUND: STIM- and Orai-mediated store operated calcium entry (SOCE) is a ubiquitous Ca2+ signaling process, crucial for the proper function of immune, muscle and neuronal systems. To treat SOCE-related disorder or diseases of these systems, and to mechanistically dissect activation and function of SOCE, specific SOCE inhibitors are needed. However, strategies for developing new SOCE modifiers are still limited.

Methodology: In this study, we identified a novel SOCE inhibitor named 2PHDO from a small pool of Chinese herbal extracts used for treating psoriasis. It could block SOCE and SOCE-mediated NFAT translocation in multiple types of cells with a half inhibitory concentration around 1 µM. At this concentration, 2PHDO was specific for SOCE. Mechanistically, 2PHDO didn't affect the activation of STIM1 or its physical coupling with Orai1. Rather, 2PHDO inhibited SOCE via its actions on Orai1.

Results: 2PHDO may serve as a good template for developing new medicines aiming to treat SOCE related diseases.

Conclusion: Overall, we proved the feasibility of screening and identification of novel SOCE inhibitors from active monomers of Chinese herbal medicine.

3.
Cell Calcium ; 112: 102735, 2023 06.
Article in English | MEDLINE | ID: mdl-37126912

ABSTRACT

The STIM-Orai signaling pathway mediates Ca2+ signals vital for controlling transcription and cell growth. The Ca2+ sensing STIM proteins are activated by depletion of Ca2+ stored in the ER, and translocate into ER-PM junctions to gate PM Orai channels. STIM1 activation also results from heating STIM1 proteins, and new evidence reveals the STIM1-mediated gating of Orai1 channels is activated by noxious cooling of cells. This activation of the STIM-Orai pathway may be important in mediating vascular dilation that occurs in response to severe cold exposure.


Subject(s)
Calcium Signaling , Signal Transduction , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Calcium Signaling/physiology , Calcium/metabolism
4.
Nat Methods ; 20(6): 918-924, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37081094

ABSTRACT

Genetically encoded calcium indicators (GECIs) are indispensable tools for real-time monitoring of intracellular calcium signals and cellular activities in living organisms. Current GECIs face the challenge of suboptimal peak signal-to-baseline ratio (SBR) with limited resolution for reporting subtle calcium transients. We report herein the development of a suite of calcium sensors, designated NEMO, with fast kinetics and wide dynamic ranges (>100-fold). NEMO indicators report Ca2+ transients with peak SBRs around 20-fold larger than the top-of-the-range GCaMP6 series. NEMO sensors further enable the quantification of absolution calcium concentration with ratiometric or photochromic imaging. Compared with GCaMP6s, NEMOs could detect single action potentials in neurons with a peak SBR two times higher and a median peak SBR four times larger in vivo, thereby outperforming most existing state-of-the-art GECIs. Given their high sensitivity and resolution to report intracellular Ca2+ signals, NEMO sensors may find broad applications in monitoring neuronal activities and other Ca2+-modulated physiological processes in both mammals and plants.


Subject(s)
Calcium , Neurons , Animals , Calcium/metabolism , Neurons/physiology , Calcium Signaling/physiology , Indicators and Reagents , Mammals/metabolism
6.
Front Pharmacol ; 14: 1111798, 2023.
Article in English | MEDLINE | ID: mdl-36817139

ABSTRACT

Introduction: Psoriasis is an inflammatory autoimmune skin disease that is hard to cure and prone to relapse. Currently available global immunosuppressive agents for psoriasis may cause severe side effects, thus it is crucial to identify new therapeutic reagents and druggable signaling pathways for psoriasis. Methods: To check the effects of SOCE inhibitors on psoriasis, we used animal models, biochemical approaches, together with various imaging techniques, including calcium, confocal and FRET imaging. Results and discussion: Store operated calcium (Ca2+) entry (SOCE), mediated by STIM1 and Orai1, is crucial for the function of keratinocytes and immune cells, the two major players in psoriasis. Here we showed that a natural compound celastrol is a novel SOCE inhibitor, and it ameliorated the skin lesion and reduced PASI scores in imiquimod-induced psoriasis-like mice. Celastrol dose- and time-dependently inhibited SOCE in HEK cells and HaCaT cells, a keratinocyte cell line. Mechanistically, celastrol inhibited SOCE via its actions both on STIM1 and Orai1. It inhibited Ca2+ entry through constitutively-active Orai1 mutants independent of STIM1. Rather than blocking the conformational switch and oligomerization of STIM1 during SOCE activation, celastrol diminished the transition from oligomerized STIM1 into aggregates, thus locking STIM1 in a partially active state. As a result, it abolished the functional coupling between STIM1 and Orai1, diminishing SOCE signals. Overall, our findings identified a new SOCE inhibitor celastrol that suppresses psoriasis, suggesting that SOCE pathway may serve as a new druggable target for treating psoriasis.

8.
iScience ; 24(11): 103339, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34816101

ABSTRACT

Transformation of naive macrophages into classically (M1) or alternatively (M2) activated macrophages regulates the inflammatory response. Here, we identified that distinct Ca2+ entry channels determine the IFNγ-induced M1 or IL-4-induced M2 transition. Naive or M2 macrophages exhibit a robust Ca2+ entry that was dependent on Orai1 channels, whereas the M1 phenotype showed a non-selective TRPC1 current. Blockade of Ca2+ entry suppresses pNF-κB/pJNK/STAT1 or STAT6 signaling events and consequently lowers cytokine production that is essential for M1 or M2 functions. Of importance, LPS stimulation shifted M2 cells from Orai1 toward TRPC1-mediated Ca2+ entry and TRPC1-/- mice exhibited transcriptional changes that suppress pro-inflammatory cytokines. In contrast, Orai1-/- macrophages showed a decrease in anti-inflammatory cytokines and exhibited a suppression of mitochondrial oxygen consumption rate and inhibited mitochondrial shape transition specifically in the M2 cells. Finally, alterations in TRPC1 or Orai1 expression determine macrophage polarization suggesting a distinct role of Ca2+ channels in modulating macrophage transformation.

9.
Cell Rep ; 35(13): 109322, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34192542

ABSTRACT

Junctional coupling between endoplasmic reticulum (ER) Ca2+-sensor STIM proteins and plasma membrane (PM) Orai channels mediates Ca2+ signals in most cells. We reveal that PM-tethered, fluorescently tagged C-terminal M4x (fourth transmembrane helix contains a cytoplasmic C-terminal extension) peptides from Orai channels undergo a Leu-specific signature of direct interaction with the STIM1 Orai-activating region (SOAR), exactly mimicking STIM1 binding to gate Orai channels. The 20-amino-acid Orai3-M4x peptide associates avidly with STIM1 within ER-PM junctions, functions to competitively block native Ca2+ signals, and mediates a key modification of STIM-Orai coupling induced by 2-aminoethoxydiphenyl borate. By blocking STIM-Orai coupling, the Orai3-M4x peptide reveals the critical role of Orai channels in driving Ca2+ oscillatory signals and transcriptional control through NFAT. The M4x peptides interact independently with SOAR dimers consistent with unimolecular coupling between Orai subunits and STIM1 dimers. We reveal the critical role of M4x helices in defining the coupling interface between STIM and Orai proteins to mediate store-operated Ca2+ signals.


Subject(s)
Calcium Signaling , ORAI1 Protein/chemistry , ORAI1 Protein/metabolism , Peptides/metabolism , Stromal Interaction Molecule 1/metabolism , Amino Acid Sequence , Binding Sites , Boron Compounds/pharmacology , Calcium Channels/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , HeLa Cells , Humans , Ion Channel Gating , Leucine/metabolism , Models, Molecular , Mutation/genetics , NFATC Transcription Factors/metabolism , Protein Binding , Protein Multimerization , Transcription, Genetic/drug effects
11.
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
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.
Elife ; 92020 09 11.
Article in English | MEDLINE | ID: mdl-32914752

ABSTRACT

Despite the established role of mitochondria in cancer, the mechanisms by which mitochondrial Ca2+ (mtCa2+) regulates tumorigenesis remain incompletely understood. The crucial role of mtCa2+ in tumorigenesis is highlighted by altered expression of proteins mediating mtCa2+ uptake and extrusion in cancer. Here, we demonstrate decreased expression of the mitochondrial Na+/Ca2+/Li+ exchanger NCLX (SLC8B1) in human colorectal tumors and its association with advanced-stage disease in patients. Downregulation of NCLX causes mtCa2+ overload, mitochondrial depolarization, decreased expression of cell-cycle genes and reduced tumor size in xenograft and spontaneous colorectal cancer mouse models. Concomitantly, NCLX downregulation drives metastatic spread, chemoresistance, and expression of epithelial-to-mesenchymal, hypoxia, and stem cell pathways. Mechanistically, mtCa2+ overload leads to increased mitochondrial reactive oxygen species, which activate HIF1α signaling supporting metastasis of NCLX-null tumor cells. Thus, loss of NCLX is a novel driver of metastasis, indicating that regulation of mtCa2+ is a novel therapeutic approach in metastatic colorectal cancer.


Colorectal cancer is the second largest cause of cancer deaths worldwide. Even in cases where the cancer is diagnosed and treated early, cells can sometimes survive treatment and spread to other organs. Once the cancer has spread, the survival rate is less than 15%. Mitochondria are compartments in the cell that produce energy, and they play an important role in supporting the rapid growth of cancer cells. The levels of calcium ions in mitochondria control how they produce energy, a process that is altered in cancer cells. To better understand how calcium ions influence colorectal cancer growth, Pathak, Gueguinou et al. studied a protein called NCLX, which controls calcium levels by pumping them out of the mitochondria. Two mouse strains that were used to study what happens if NCLX is missing. The first strain was genetically modified to disable the gene for NCLX and then exposed to carcinogens. The second strain was injected with colorectal cancer cells from a human tumor that were lacking NCLX. In both strains, the tumors that formed were smaller than in mice with NCLX. However, the human cancer cells in the second model were more likely to spread to other organs. This is likely because the build-up of calcium ions in the mitochondria of mice lacking NCLX led to an increase in the production of hypoxia-inducible factor-1a, a protein that is a common driver of cancer spread. Pathak, Gueguinou et al. demonstrated how NCLX can affect colorectal cancer progression. It suggests that it may have opposing effects during early and late-stage colorectal cancer, encouraging tumor growth but also decreasing the spread to other organs. Further research could help refine treatments at different stages of the disease.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Mitochondrial Proteins/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Calcium/metabolism , Colon/pathology , Female , Humans , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Neoplasm Metastasis
14.
Curr Opin Physiol ; 17: 106-114, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32954113

ABSTRACT

Store-operated Ca2+ entry signals are critical for cellular regulation. This intricate signaling pathway involves coupling of proteins in two different membranes: the ER-resident Ca2+-sensing STIM proteins and the highly Ca2+-selective PM Orai channels. The molecular nature of the STIM-Orai coupling interface in ER-PM junctions and consequent Orai channel gating, are processes under intense study. We describe recent developments in determining the mechanism of Orai activation through the key STIM-Orai Activating Region (SOAR) of STIM1. We describe the unexpected unimolecular coupling of STIM with Orai and explain the observed variable stoichiometry of STIM-Orai interactions. We also define the discrete C-terminal regions in Orai channels that initially latch onto STIM proteins and mediate allosteric activation of the channel. A critical "nexus" region closely associated with the STIM-activated C-terminus of Orai1, propagates the STIM-binding signal through the four tightly-associated transmembrane helices of Orai1, finally to modify the pore-forming helices and effect channel opening.

15.
Proc Natl Acad Sci U S A ; 117(29): 17369-17380, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32641503

ABSTRACT

Voltage-gated L-type Ca2+ channel (Cav1.2) blockers (LCCBs) are major drugs for treating hypertension, the preeminent risk factor for heart failure. Vascular smooth muscle cell (VSMC) remodeling is a pathological hallmark of chronic hypertension. VSMC remodeling is characterized by molecular rewiring of the cellular Ca2+ signaling machinery, including down-regulation of Cav1.2 channels and up-regulation of the endoplasmic reticulum (ER) stromal-interacting molecule (STIM) Ca2+ sensor proteins and the plasma membrane ORAI Ca2+ channels. STIM/ORAI proteins mediate store-operated Ca2+ entry (SOCE) and drive fibro-proliferative gene programs during cardiovascular remodeling. SOCE is activated by agonists that induce depletion of ER Ca2+, causing STIM to activate ORAI. Here, we show that the three major classes of LCCBs activate STIM/ORAI-mediated Ca2+ entry in VSMCs. LCCBs act on the STIM N terminus to cause STIM relocalization to junctions and subsequent ORAI activation in a Cav1.2-independent and store depletion-independent manner. LCCB-induced promotion of VSMC remodeling requires STIM1, which is up-regulated in VSMCs from hypertensive rats. Epidemiology showed that LCCBs are more associated with heart failure than other antihypertensive drugs in patients. Our findings unravel a mechanism of LCCBs action on Ca2+ signaling and demonstrate that LCCBs promote vascular remodeling through STIM-mediated activation of ORAI. Our data indicate caution against the use of LCCBs in elderly patients or patients with advanced hypertension and/or onset of cardiovascular remodeling, where levels of STIM and ORAI are elevated.


Subject(s)
Calcium Channels, L-Type/metabolism , Hypertension/metabolism , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/metabolism , Stromal Interaction Molecules/metabolism , Vascular Remodeling/physiology , Animals , Antihypertensive Agents/pharmacology , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Gene Knockout Techniques , HEK293 Cells , Heart Failure , Humans , Membrane Proteins/genetics , Myocytes, Smooth Muscle , Neoplasm Proteins , ORAI1 Protein/genetics , Rats , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 2/genetics
16.
Nat Commun ; 11(1): 2444, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415068

ABSTRACT

The essential role of ORAI1 channels in receptor-evoked Ca2+ signaling is well understood, yet little is known about the physiological activation of the ORAI channel trio natively expressed in all cells. The roles of ORAI2 and ORAI3 have remained obscure. We show that ORAI2 and ORAI3 channels play a critical role in mediating the regenerative Ca2+ oscillations induced by physiological receptor activation, yet ORAI1 is dispensable in generation of oscillations. We reveal that ORAI2 and ORAI3 channels multimerize with ORAI1 to expand the range of sensitivity of receptor-activated Ca2+ signals, reflecting their enhanced basal STIM1-binding and heightened Ca2+-dependent inactivation. This broadened bandwidth of Ca2+ influx is translated by cells into differential activation of NFAT1 and NFAT4 isoforms. Our results uncover a long-sought role for ORAI2 and ORAI3, revealing an intricate control mechanism whereby heteromerization of ORAI channels mediates graded Ca2+ signals that extend the agonist-sensitivity to fine-tune transcriptional control.


Subject(s)
Calcium Release Activated Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/drug effects , Carbachol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Models, Biological , NFATC Transcription Factors/metabolism , ORAI1 Protein/metabolism , Protein Binding/drug effects , Protein Isoforms/metabolism , Protein Multimerization/drug effects , Stromal Interaction Molecule 1/metabolism , Time-Lapse Imaging
17.
Cells ; 9(2)2020 01 22.
Article in English | MEDLINE | ID: mdl-31979185

ABSTRACT

Being the largest the Ca2+ store in mammalian cells, endoplasmic reticulum (ER)-mediated Ca2+ signalling often involves both Ca2+ release via inositol 1, 4, 5-trisphosphate receptors (IP3R) and store operated Ca2+ entries (SOCE) through Ca2+ release activated Ca2+ (CRAC) channels on plasma membrane (PM). IP3Rs are functionally coupled with CRAC channels and other Ca2+ handling proteins. However, it still remains less well defined as to whether IP3Rs could regulate ER-mediated Ca2+ signals independent of their Ca2+ releasing ability. To address this, we generated IP3Rs triple and double knockout human embryonic kidney (HEK) cell lines (IP3Rs-TKO, IP3Rs-DKO), and systemically examined ER Ca2+ dynamics and CRAC channel activity in these cells. The results showed that the rate of ER Ca2+ leakage and refilling, as well as SOCE were all significantly reduced in IP3Rs-TKO cells. And these TKO effects could be rescued by over-expression of IP3R3. Further, results showed that the diminished SOCE was caused by NEDD4L-mediated ubiquitination of Orai1 protein. Together, our findings indicate that IP3R3 is one crucial player in coordinating ER-mediated Ca2+ signalling.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Calcium Signaling , Cell Movement , Cell Proliferation , HEK293 Cells , Humans , Nedd4 Ubiquitin Protein Ligases/metabolism , ORAI1 Protein/metabolism , Protein Isoforms/metabolism
18.
PLoS Biol ; 17(8): e3000413, 2019 08.
Article in English | MEDLINE | ID: mdl-31469825

ABSTRACT

Calcium signals drive an endless array of cellular responses including secretion, contraction, transcription, cell division, and growth. The ubiquitously expressed Orai family of plasma membrane (PM) ion channels mediate Ca2+ entry signals triggered by the Ca2+ sensor Stromal Interaction Molecule (STIM) proteins of the endoplasmic reticulum (ER). The 2 proteins interact within curiously obscure ER-PM junctions, driving an allosteric gating mechanism for the Orai channel. Although key to Ca2+ signal generation, molecular understanding of this activation process remain obscure. Crystallographic structural analyses reveal much about the exquisite hexameric core structure of Orai channels. But how STIM proteins bind to the channel periphery and remotely control opening of the central pore, has eluded such analysis. Recent studies apply both crystallography and single-particle cryogenic electron microscopy (cryo-EM) analyses to probe the structure of Orai mutants that mimic activation by STIM. The results provide new understanding on the open state of the channel and how STIM proteins may exert remote allosteric control of channel gating.


Subject(s)
Calcium Channels , Calcium , Calcium Signaling , ORAI1 Protein , Stromal Interaction Molecule 1
19.
Nat Commun ; 10(1): 1971, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036819

ABSTRACT

ORAI1 constitutes the store-operated Ca2+ release-activated Ca2+ (CRAC) channel crucial for life. Whereas ORAI1 activation by Ca2+-sensing STIM proteins is known, still obscure is how ORAI1 is turned off through Ca2+-dependent inactivation (CDI), protecting against Ca2+ toxicity. Here we identify a spatially-restricted Ca2+/cAMP signaling crosstalk critical for mediating CDI. Binding of Ca2+-activated adenylyl cyclase 8 (AC8) to the N-terminus of ORAI1 positions AC8 near the mouth of ORAI1 for sensing Ca2+. Ca2+ permeating ORAI1 activates AC8 to generate cAMP and activate PKA. PKA, positioned by AKAP79 near ORAI1, phosphorylates serine-34 in ORAI1 pore extension to induce CDI whereas recruitment of the phosphatase calcineurin antagonizes the effect of PKA. Notably, CDI shapes ORAI1 cytosolic Ca2+ signature to determine the isoform and degree of NFAT activation. Thus, we uncover a mechanism of ORAI1 inactivation, and reveal a hitherto unappreciated role for inactivation in shaping cellular Ca2+ signals and NFAT activation.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , ORAI1 Protein/metabolism , A Kinase Anchor Proteins/metabolism , Blotting, Western , Cyclic AMP-Dependent Protein Kinases/metabolism , HEK293 Cells , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/genetics , Phosphorylation , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/genetics , Stromal Interaction Molecule 2/metabolism
20.
SELECTION OF CITATIONS
SEARCH DETAIL
...