Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 152(4): 778-790, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23394946

ABSTRACT

Survival in the wild requires organismal adaptations to the availability of nutrients. Endosomes and lysosomes are key intracellular organelles that couple nutrition and metabolic status to cellular responses, but how they detect cytosolic ATP levels is not well understood. Here, we identify an endolysosomal ATP-sensitive Na(+) channel (lysoNa(ATP)). The channel is a complex formed by two-pore channels (TPC1 and TPC2), ion channels previously thought to be gated by nicotinic acid adenine dinucleotide phosphate (NAADP), and the mammalian target of rapamycin (mTOR). The channel complex detects nutrient status, becomes constitutively open upon nutrient removal and mTOR translocation off the lysosomal membrane, and controls the lysosome's membrane potential, pH stability, and amino acid homeostasis. Mutant mice lacking lysoNa(ATP) have much reduced exercise endurance after fasting. Thus, TPCs make up an ion channel family that couples the cell's metabolic state to endolysosomal function and are crucial for physical endurance during food restriction.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium Channels/metabolism , Lysosomes/metabolism , Sodium Channels/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenylate Kinase/metabolism , Amino Acids/metabolism , Animals , Calcium Channels/chemistry , Calcium Channels/genetics , Fasting , Gene Knockout Techniques , Homeostasis , Humans , Hydrogen-Ion Concentration , Membrane Potentials , Mice , Physical Endurance
2.
Cell ; 151(2): 372-83, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063126

ABSTRACT

Mammalian two-pore channel proteins (TPC1, TPC2; TPCN1, TPCN2) encode ion channels in intracellular endosomes and lysosomes and were proposed to mediate endolysosomal calcium release triggered by the second messenger, nicotinic acid adenine dinucleotide phosphate (NAADP). By directly recording TPCs in endolysosomes from wild-type and TPC double-knockout mice, here we show that, in contrast to previous conclusions, TPCs are in fact sodium-selective channels activated by PI(3,5)P(2) and are not activated by NAADP. Moreover, the primary endolysosomal ion is Na(+), not K(+), as had been previously assumed. These findings suggest that the organellar membrane potential may undergo large regulatory changes and may explain the specificity of PI(3,5)P(2) in regulating the fusogenic potential of intracellular organelles.


Subject(s)
Calcium Channels/metabolism , Lysosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Knockout , NADP/analogs & derivatives , NADP/metabolism , Sodium Channels/metabolism
3.
Mol Cell ; 58(2): 197-9, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25884365

ABSTRACT

In this issue of Molecular Cell, Kar and Parekh (2015) reveal the remarkable intricacy and accuracy of Ca(2+) signals in differentially controlling the function of closely related transcription factors.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , NFATC Transcription Factors/metabolism , Humans
4.
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
5.
Clin Otolaryngol ; 47(6): 664-671, 2022 11.
Article in English | MEDLINE | ID: mdl-36073732

ABSTRACT

OBJECTIVE: To investigate the associations between weekly alcohol consumption and the risk and surgical outcome of Chronic Rhinosinusitis (CRS). DESIGN: A case-control study. SETTING AND PARTICIPANTS: The study population consisted of 1095 CRS patients and 909 healthy collected from the first affiliated hospital of Zhengzhou University between May 2018 and December 2019. MAIN OUTCOME MEASURES: Unconditional multivariate logistic regression analysis and Cox proportional hazards regression analysis were performed to estimate the association of alcohol consumption with the risk and surgical outcomes of CRS. Odds ratios (OR) or hazard ratio (HR) with 95% confidence intervals (CI) were calculated separately. The Kruskal-Wallis test was used to explore the possible molecular mechanisms. RESULTS: As compared with nondrinkers, the multivariable-adjusted OR (95% CI) values of current drinkers consuming 7.5-22 drinks and >22 drinks per week were 2.158 (1.249-3.729) and 5.373 (2.912-9.911), respectively. The rate of mucosal epithelialization after CRS surgery for patients who drank 7.5-22 drinks and >22 drinks per week was lower than that of nondrinkers [HR (95% CI) = 0.487 (0.351-0.675) and 0.252 (0.184-0.346), respectively]. The association of alcohol consumption with the risk and surgical outcome of CRS was dose dependent (p < .01). Alcohol consumption increased the risk of CRS and extended the time of mucosal epithelialization after CRS surgery by possibly increasing serum IgE levels (p < .05). CONCLUSION: Higher alcohol consumption of >7.5 drinks per week was an independent risk factor for CRS and extended the time of mucosal epithelialization after surgery. As a potential underlying mechanism, alcohol consumption increases serum IgE levels.


Subject(s)
Alcohol Drinking , Immunoglobulin E , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Case-Control Studies , China/epidemiology , Humans , Prospective Studies , Risk Factors , Treatment Outcome
6.
EMBO J ; 36(6): 797-815, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28219928

ABSTRACT

Mitochondria exert important control over plasma membrane (PM) Orai1 channels mediating store-operated Ca2+ entry (SOCE). Although the sensing of endoplasmic reticulum (ER) Ca2+ stores by STIM proteins and coupling to Orai1 channels is well understood, how mitochondria communicate with Orai1 channels to regulate SOCE activation remains elusive. Here, we reveal that SOCE is accompanied by a rise in cytosolic Na+ that is critical in activating the mitochondrial Na+/Ca2+ exchanger (NCLX) causing enhanced mitochondrial Na+ uptake and Ca2+ efflux. Omission of extracellular Na+ prevents the cytosolic Na+ rise, inhibits NCLX activity, and impairs SOCE and Orai1 channel current. We show further that SOCE activates a mitochondrial redox transient which is dependent on NCLX and is required for preventing Orai1 inactivation through oxidation of a critical cysteine (Cys195) in the third transmembrane helix of Orai1. We show that mitochondrial targeting of catalase is sufficient to rescue redox transients, SOCE, and Orai1 currents in NCLX-deficient cells. Our findings identify a hitherto unknown NCLX-mediated pathway that coordinates Na+ and Ca2+ signals to effect mitochondrial redox control over SOCE.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , ORAI1 Protein/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium/metabolism , Cell Line , Humans , Mitochondrial Proteins , Oxidation-Reduction
7.
Proc Natl Acad Sci U S A ; 115(15): E3398-E3407, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581306

ABSTRACT

The transmembrane docking of endoplasmic reticulum (ER) Ca2+-sensing STIM proteins with plasma membrane (PM) Orai Ca2+ channels is a critical but poorly understood step in Ca2+ signal generation. STIM1 protein dimers unfold to expose a discrete STIM-Orai activating region (SOAR1) that tethers and activates Orai1 channels within discrete ER-PM junctions. We reveal that each monomer within the SOAR dimer interacts independently with single Orai1 subunits to mediate cross-linking between Orai1 channels. Superresolution imaging and mobility measured by fluorescence recovery after photobleaching reveal that SOAR dimer cross-linking leads to substantial Orai1 channel clustering, resulting in increased efficacy and cooperativity of Orai1 channel function. A concatenated SOAR1 heterodimer containing one monomer point mutated at its critical Orai1 binding residue (F394H), although fully activating Orai channels, is completely defective in cross-linking Orai1 channels. Importantly, the naturally occurring STIM2 variant, STIM2.1, has an eight-amino acid insert in its SOAR unit that renders it functionally identical to the F394H mutant in SOAR1. Contrary to earlier predictions, the SOAR1-SOAR2.1 heterodimer fully activates Orai1 channels but prevents cross-linking and clustering of channels. Interestingly, combined expression of full-length STIM1 with STIM2.1 in a 5:1 ratio causes suppression of sustained agonist-induced Ca2+ oscillations and protects cells from Ca2+ overload, resulting from high agonist-induced Ca2+ release. Thus, STIM2.1 exerts a powerful regulatory effect on signal generation likely through preventing Orai1 channel cross-linking. Overall, STIM-mediated cross-linking of Orai1 channels is a hitherto unrecognized functional paradigm that likely provides an organizational microenvironment within ER-PM junctions with important functional impact on Ca2+ signal generation.


Subject(s)
Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , ORAI1 Protein/chemistry , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/chemistry , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/chemistry , Stromal Interaction Molecule 2/metabolism , Calcium/metabolism , Dimerization , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , Protein Domains , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 2/genetics
8.
J Biol Chem ; 294(16): 6318-6332, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30824535

ABSTRACT

Store-operated Ca2+ entry (SOCE) is a ubiquitous pathway for Ca2+ influx across the plasma membrane (PM). SOCE is mediated by the endoplasmic reticulum (ER)-associated Ca2+-sensing proteins stromal interaction molecule 1 (STIM1) and STIM2, which transition into an active conformation in response to ER Ca2+ store depletion, thereby interacting with and gating PM-associated ORAI1 channels. Although structurally homologous, STIM1 and STIM2 generate distinct Ca2+ signatures in response to varying strengths of agonist stimulation. The physiological functions of these Ca2+ signatures, particularly under native conditions, remain unclear. To investigate the structural properties distinguishing STIM1 and STIM2 activation of ORAI1 channels under native conditions, here we used CRISPR/Cas9 to generate STIM1-/-, STIM2-/-, and STIM1/2-/- knockouts in HEK293 and colorectal HCT116 cells. We show that depending on cell type, STIM2 can significantly sustain SOCE in response to maximal store depletion. Utilizing the SOCE modifier 2-aminoethoxydiphenyl borate (2-APB), we demonstrate that 2-APB-activated store-independent Ca2+ entry is mediated exclusively by endogenous STIM2. Using variants that either stabilize or disrupt intramolecular interactions of STIM C termini, we show that the increased flexibility of the STIM2 C terminus contributes to its selective store-independent activation by 2-APB. However, STIM1 variants with enhanced flexibility in the C terminus failed to support its store-independent activation. STIM1/STIM2 chimeric constructs indicated that coordination between N-terminal sensitivity and C-terminal flexibility is required for specific store-independent STIM2 activation. Our results clarify the structural determinants underlying activation of specific STIM isoforms, insights that are potentially useful for isoform-selective drug targeting.


Subject(s)
Calcium Signaling , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Stromal Interaction Molecule 2/metabolism , Boron Compounds/chemistry , Boron Compounds/pharmacology , Calcium/chemistry , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/genetics , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , Humans , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Stromal Interaction Molecule 1/chemistry , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/chemistry , Stromal Interaction Molecule 2/genetics
9.
J Cell Mol Med ; 23(3): 1687-1697, 2019 03.
Article in English | MEDLINE | ID: mdl-30636376

ABSTRACT

Tissue damage and its associated-inflammation act as tumour initiators or propagators. AMP-activated protein kinase (AMPK) is activated by environmental or nutritional stress factors, such as hypoxia, glucose deprivation, and other cell injury factors, to regulate cell energy balance and differentiation. We previously have reported that AMPKα2 deficiency resulted in the energy deprivation in tumour-bearing liver and the enhanced-hepatocyte death. In this study, AMPKα2 knockout mice and the liver metastasis model of colon cancer cells were used to address the role of AMPKα isoforms in tumour inflammation. First, we found that the AMPKα2 deficiency exacerbated the liver injury and recruitment of macrophages. Meanwhile, although compensatory expression of AMPKα1 was not significant after AMPKα2 knockout, AMPKα1 phosphorylation was elevated in remnant liver in AMPKα2 knockout mice, which was positively associated with the enhanced energy deprivation in the AMPKα2 deficient mice. Furthermore, the activated AMPKα1 in macrophage contributed to its polarizing to tumour-associated phenotype. Thus, the enhanced tumour-associated inflammation and activation of AMPKα1 in the AMPKα2 deficient mice may exacerbate the tumour development by affecting the tumour inflammatory microenvironment. Our study suggests that the two isoforms of AMPKα, AMPKα1 and AMPKα2 play different roles in controlling tumour development.


Subject(s)
AMP-Activated Protein Kinases/physiology , Colonic Neoplasms/etiology , Disease Models, Animal , Inflammation/etiology , Liver Neoplasms/etiology , Macrophages/pathology , Animals , Cell Differentiation , Cells, Cultured , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Energy Metabolism , Inflammation/metabolism , Inflammation/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Phosphorylation , Tumor Microenvironment
10.
J Biol Chem ; 293(33): 12962-12974, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29954946

ABSTRACT

Store-operated Ca2+ entry signals are mediated by plasma membrane Orai channels activated through intermembrane coupling with Ca2+-sensing STIM proteins in the endoplasmic reticulum (ER). The nature of this elaborate Orai-gating mechanism has remained enigmatic. Based on the Drosophila Orai structure, mammalian Orai1 channels are hexamers comprising three dimeric subunit pairs. We utilized concatenated Orai1 dimers to probe the function of key domains within the channel pore and gating regions. The Orai1-E106Q selectivity-filter mutant, widely considered a dominant pore blocker, was surprisingly nondominant within concatenated heterodimers with Orai1-WT. The Orai1-E106Q/WT heterodimer formed STIM1-activated nonselective cation channels with significantly enlarged apparent pore diameter. Other Glu-106 substitutions entirely blocked the function of heterodimers with Orai1-WT. The hydrophobic pore-lining mutation V102C, which constitutively opens channels, was suppressed by Orai1-WT in the heterodimer. In contrast, the naturally occurring R91W pore-lining mutation associated with human immunodeficiency was completely dominant-negative over Orai-WT in heterodimers. Heterodimers containing the inhibitory K85E mutation extending outward from the pore helix gave an interesting partial effect on both channel activation and STIM1 binding, indicating an important allosteric link between the cytosolic Orai1 domains. The Orai1 C-terminal STIM1-binding domain mutation L273D powerfully blocked STIM1-induced channel activation. The Orai1-L273D/WT heterodimer had drastically impaired STIM1-induced channel gating but, unexpectedly, retained full STIM1 binding. This reveals the critical role of Leu-273 in transducing the STIM1-binding signal into the allosteric conformational change that initiates channel gating. Overall, our results provide important new insights into the role of key functional domains that mediate STIM1-induced gating of the Orai1 channel.


Subject(s)
Endoplasmic Reticulum/metabolism , Ion Channel Gating , Mutation, Missense , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , Protein Multimerization , Stromal Interaction Molecule 1/metabolism , Allosteric Regulation , Amino Acid Substitution , Animals , Drosophila melanogaster , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , Protein Domains , Stromal Interaction Molecule 1/genetics
11.
Pflugers Arch ; 470(10): 1555-1567, 2018 10.
Article in English | MEDLINE | ID: mdl-29934936

ABSTRACT

Mediated through the combined action of STIM proteins and Orai channels, store-operated Ca2+ entry (SOCE) functions ubiquitously among different cell types. The existence of multiple STIM and Orai genes has made it difficult to assign specific roles of each STIM and Orai homolog in mediating Ca2+ signals. Using CRISPR/Cas9 gene editing tools, we generated cells with both STIM or all three Orai homologs deleted and directly monitored store Ca2+ and Ca2+ signals. We found that unstimulated, SOCE null KO cells still retain 50~70% of ER Ca2+ stores of wildtype (wt) cells. After brief exposure to store-emptying conditions, acute refilling of ER Ca2+ stores was totally blocked in KO cells. However, after 24 h in culture, stores were eventually refilled. Thus, SOCE is critical for immediate refilling of ER Ca2+ but is dispensable for the maintenance of long-term ER Ca2+ homeostasis. Using the Orai null background triple Orai-KO cells, we examined the plasma membrane translocation properties of a series of truncated STIM1 variants. FRET analysis reveals that, even though PM tethering of STIM1 expedites the activation of STIM1 by facilitating its oligomerization, migration, and accumulation in ER-PM junctions, it is not required for the conformational switch, oligomerization, and clustering of STIM1. Even without overt puncta formation at ER-PM junctions, STIM11-491 and STIM11-666 could still rescue SOCE when expressed in STIM KO cells. Thus, ER-PM trapping and clustering of STIM molecules only facilitates the process of SOCE activation, but is not essential for the activation of Orai channels.


Subject(s)
Calcium Signaling , ORAI1 Protein/deficiency , Stromal Interaction Molecule 1/deficiency , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , ORAI1 Protein/genetics , Protein Multimerization , Protein Transport , Stromal Interaction Molecule 1/genetics
12.
J Biol Chem ; 291(50): 25764-25775, 2016 Dec 09.
Article in English | MEDLINE | ID: mdl-27780862

ABSTRACT

Orai channels mediate store-operated Ca2+ signals crucial in regulating transcription in many cell types, and implicated in numerous immunological and inflammatory disorders. Despite their central importance, controversy surrounds the basic subunit structure of Orai channels, with several biochemical and biophysical studies suggesting a tetrameric structure yet crystallographic evidence indicating a hexamer. We systematically investigated the subunit configuration of the functional Orai1 channel, generating a series of tdTomato-tagged concatenated Orai1 channel constructs (dimers to hexamers) expressed in CRISPR-derived ORAI1 knock-out HEK cells, stably expressing STIM1-YFP. Surface biotinylation demonstrated that the full-length concatemers were surface membrane-expressed. Unexpectedly, Orai1 dimers, trimers, tetramers, pentamers, and hexamers all mediated similar and substantial store-operated Ca2+ entry. Moreover, each Orai1 concatemer mediated Ca2+ currents with inward rectification and reversal potentials almost identical to those observed with expressed Orai1 monomer. In Orai1 tetramers, subunit-specific replacement with Orai1 E106A "pore-inactive" subunits revealed that functional channels utilize only the N-terminal dimer from the tetramer. In contrast, Orai1 E106A replacement in Orai1 hexamers established that all the subunits can contribute to channel formation, indicating a hexameric channel configuration. The critical Ca2+ selectivity filter-forming Glu-106 residue may mediate Orai1 channel assembly around a central Ca2+ ion within the pore. Thus, multiple E106A substitutions in the Orai1 hexamer may promote an alternative "trimer-of-dimers" channel configuration in which the C-terminal E106A subunits are excluded from the hexameric core. Our results argue strongly against a tetrameric configuration for Orai1 channels and indicate that the Orai1 channel functions as a hexamer.


Subject(s)
Calcium , ORAI1 Protein/metabolism , Protein Multimerization/physiology , Amino Acid Substitution , Gene Knockdown Techniques , HEK293 Cells , Humans , Mutation, Missense , ORAI1 Protein/genetics
13.
Adv Exp Med Biol ; 993: 83-98, 2017.
Article in English | MEDLINE | ID: mdl-28900910

ABSTRACT

Store-operated Ca2+ entry fulfills a crucial role in controlling Ca2+ signals in almost all cells. The Ca2+-sensing stromal interaction molecule (STIM) proteins in the endoplasmic reticulum (ER) undergo complex conformational changes in response to depleted ER luminal Ca2+, allowing them to unfold and become trapped in ER-plasma membrane (PM) junctions. Dimers of STIM proteins trap and gate the plasma membrane Orai Ca2+ channels within these junctions to generate discrete zones of high Ca2+ and regulate sensitive Ca2+-dependent intracellular signaling pathways. The STIM-Orai activating region (SOAR) of STIM1 becomes exposed upon store depletion and promotes trapping of Orai1 at the PM. Residue Phe-394 within SOAR forms an integral part of the high-affinity Orai1-interacting site. Our results demonstrate that only a single active site within the dimeric SOAR domain of STIM1 is required for the activation of Orai1 channel activity. This unimolecular model is strongly supported by evidence of variable STIM1:Orai1 stoichiometry reported in many studies. We hypothesize that unimolecular coupling promotes cross-linking of channels, localizing Ca2+ signals, and regulating channel activity. We have also identified a key "nexus" region in Orai1 near the C-terminal STIM1-binding site that can be mutated to constitutively activate Ca2+ entry, mimicking STIM1 activated channels. This suggests that STIM1 mediates gating of Orai1 in an allosteric manner via interaction with the Orai1 C-terminus alone. This model suggests the dual role of STIM1 in regulating both localization and gating of Orai1 channels and has important implications for the regulation of SOCE-mediated downstream signaling and the kinetics of channel activation.


Subject(s)
Calcium/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Calcium Signaling/physiology , Cell Membrane/metabolism , Humans
14.
Pflugers Arch ; 468(11-12): 2061-2074, 2016 11.
Article in English | MEDLINE | ID: mdl-27726010

ABSTRACT

Store-operated Ca2+ entry (SOCE) mediated by STIM1 and Orai1 is crucial for Ca2+ signaling and homeostasis in most cell types. 2-Aminoethoxydiphenyl borate (2-APB) is a well-described SOCE inhibitor, but its mechanisms of action remain largely elusive. Here, we show that 2-APB does not affect the dimeric state of STIM1, but enhances the intramolecular coupling between the coiled-coil 1 (CC1) and STIM-Orai-activating region (SOAR) of STIM1, with subsequent reduction in the formation of STIM1 puncta in the absence of Orai1 overexpression. 2-APB also inhibits Orai1 channels, directly inhibiting Ca2+ entry through the constitutively active, STIM1-independent Orai1 mutants, Orai1-P245T and Orai1-V102A. When unbound from STIM1, the constitutively active Orai1-V102C mutant is not inhibited by 2-APB. Thus, we used Orai1-V012C as a tool to examine whether 2-APB can also inhibit the coupling between STIM1 and Orai1. We reveal that the functional coupling between STIM1 and Orai1-V102C is inhibited by 2-APB. This inhibition on coupling is indirect, arising from 2-APB's action on STIM1, and it is most likely mediated by functional channel residues in the Orai1 N-terminus. Overall, our findings on this two-site inhibition mediated by 2-APB provide new understanding on Orai1-activation by STIM1, important to future drug design.


Subject(s)
Calcium Signaling/drug effects , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Boron Compounds/pharmacology , Calcium/metabolism , HEK293 Cells , Humans
15.
Asian J Androl ; 26(4): 366-376, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38738948

ABSTRACT

For sperm cryopreservation, the conventional method, which requires glycerol, has been used for a long time. In addition, the permeable cryoprotectant-free vitrification method has been continuously studied. Although the differences of cryopreservation effects between the two methods have being studied, differences in microRNA (miRNA) profiles between them remain unclear. In this study, we investigated the differences in miRNA expression profiles among conventional freezing sperm, droplet vitrification freezing sperm and fresh human sperm. We also analyzed the differences between these methods in terms of differentially expressed miRNAs (DEmiRs) related to early embryonic development and paternal epigenetics. Our results showed no significant differences between the cryopreservation methods in terms of sperm motility ratio, plasma membrane integrity, DNA integrity, mitochondrial membrane potential, acrosome integrity, and ultrastructural damage. However, sperm miRNA-sequencing showed differences between the two methods in terms of the numbers of DEmiRs (28 and 19 with vitrification using a nonpermeable cryoprotectant and the conventional method, respectively) in postthaw and fresh sperm specimens. DEmiRs related to early embryonic development and paternal epigenetics mainly included common DEmiRs between the groups. Our results showed that the differences between conventional freezing and droplet vitrification were minimal in terms of miRNA expression related to embryonic development and epigenetics. Changes in sperm miRNA expression due to freezing are not always detrimental to embryonic development. This study compared differences in miRNA expression profiles before and after cryopreservation between cryopreservation by conventional and vitrification methods. It offers a new perspective to evaluate various methods of sperm cryopreservation.


Subject(s)
Cryopreservation , MicroRNAs , Semen Preservation , Spermatozoa , Vitrification , Humans , Male , Cryopreservation/methods , MicroRNAs/genetics , Spermatozoa/metabolism , Semen Preservation/methods , Cryoprotective Agents/pharmacology , Sperm Motility/genetics , Freezing
16.
Cancers (Basel) ; 15(9)2023 Apr 23.
Article in English | MEDLINE | ID: mdl-37173886

ABSTRACT

Cell-cell communication, either through direct contact or indirectly, is critical for multiple cellular processes, such as proliferation, survival, differentiation, and transdifferentiation, and it plays a fundamental role in maintaining the integrity of tissue structure and cellular environment [...].

17.
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
18.
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.

19.
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.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL