Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Publication year range
1.
J Cell Physiol ; 238(9): 2050-2062, 2023 09.
Article in English | MEDLINE | ID: mdl-37332264

ABSTRACT

Orai1 is the pore-forming subunit of the store-operated Ca2+ release-activated Ca2+ (CRAC) channels involved in a variety of cellular functions. Two Orai1 variants have been identified, the long form, Orai1α, containing 301 amino acids, and the short form, Orai1ß, which arises from alternative translation initiation from methionines 64 or 71, in Orai1α. Orai1 is mostly expressed in the plasma membrane, but a subset of Orai1 is located in intracellular compartments. Here we show that Ca2+ store depletion leads to trafficking and insertion of compartmentalized Orai1α in the plasma membrane via a mechanism that is independent on changes in cytosolic free-Ca2+ concentration, as demonstrated by cell loading with the fast intracellular Ca2+ chelator dimethyl BAPTA in the absence of extracellular Ca2+ . Interestingly, thapsigargin (TG) was found to be unable to induce translocation of Orai1ß to the plasma membrane when expressed individually; by contrast, when Orai1ß is co-expressed with Orai1α, cell treatment with TG induced rapid trafficking and insertion of compartmentalized Orai1ß in the plasma membrane. Translocation of Orai1 forms to the plasma membrane was found to require the integrity of the actin cytoskeleton. Finally, expression of a dominant negative mutant of the small GTPase ARF6, and ARF6-T27N, abolished the translocation of compartmentalized Orai1 variants to the plasma membrane upon store depletion. These findings provide new insights into the mechanism that regulate the plasma membrane abundance of Orai1 variants after Ca2+ store depletion.


Subject(s)
Calcium Channels , Calcium Release Activated Calcium Channels , ORAI1 Protein , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Release Activated Calcium Channels/metabolism , Calcium Signaling , Cell Membrane/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Thapsigargin/pharmacology , Humans , HEK293 Cells
2.
J Physiol ; 600(7): 1631-1650, 2022 04.
Article in English | MEDLINE | ID: mdl-35081662

ABSTRACT

Regardless of its aetiology, sustained intracellular Ca2+ overload is a well-known hallmark of acute pancreatitis (AP). Toxic Ca2+ elevation induces pancreatic ductal cell damage characterized by impaired ion and fluid secretion - essential to wash out the protein-rich fluid secreted by acinar cells while maintaining the alkaline intra-ductal pH under physiological conditions - and mitochondrial dysfunction. While prevention of ductal cell injury decreases the severity of AP, no specific drug target has yet been identified in the ductal cells. Although Orai1, a store-operated Ca2+ influx channel, is known to contribute to sustained Ca2+ overload in acinar cells, details concerning its expression and function in ductal cells are currently lacking. In this study, we demonstrate that functionally active Orai1 channels reside predominantly in the apical plasma membrane of pancreatic ductal cells. Selective CM5480-mediated Orai1 inhibition impairs Stim1-dependent extracellular Ca2+ influx evoked by bile acids or ethanol combined with non-oxidative ethanol metabolites. Furthermore, prevention of sustained extracellular Ca2+ influx protects ductal cell secretory function in vitro and decreases pancreatic ductal cell death. Finally, Orai1 inhibition partially restores and maintains proper exocrine pancreatic secretion in in vivo AP models. In conclusion, our results indicate that Orai1 inhibition prevents AP-related ductal cell function impairment and holds the potential of improving disease outcome. KEY POINTS: Sustained intracellular Ca2+ overload in pancreatic acinar and ductal cells is a hallmark of biliary and alcohol-induced acute pancreatitis, which leads to impaired ductal ion and fluid secretion. Orai1 is a plasma membrane Ca2+ channel that mediates extracellular Ca2+ influx upon endoplasmic reticulum Ca2+ depletion. Results showed that Orai1 is expressed on the luminal plasma membrane of the ductal cells and selective Orai1 inhibition impaired Stim1-dependent extracellular Ca2+ influx evoked by bile acids or ethanol combined with non-oxidative ethanol metabolites. The prevention of sustained extracellular Ca2+ influx protected ductal cell secretory functions in in vitro models and maintained exocrine pancreatic secretion in in vivo acute pancreatitis models. Orai1 inhibition prevents the bile acid- and alcohol-induced damage of the pancreatic ductal secretion and holds the potential of improving the outcome of acute pancreatitis.


Subject(s)
Pancreatitis , Acute Disease , Bile Acids and Salts/toxicity , Calcium/metabolism , Calcium Signaling , Ethanol/toxicity , Humans , ORAI1 Protein/antagonists & inhibitors , Pancreatitis/drug therapy , Pancreatitis/etiology , Pancreatitis/metabolism , Stromal Interaction Molecule 1/metabolism
3.
Circulation ; 141(3): 199-216, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31906693

ABSTRACT

BACKGROUND: Orai1 is a critical ion channel subunit, best recognized as a mediator of store-operated Ca2+ entry (SOCE) in nonexcitable cells. SOCE has recently emerged as a key contributor of cardiac hypertrophy and heart failure but the relevance of Orai1 is still unclear. METHODS: To test the role of these Orai1 channels in the cardiac pathophysiology, a transgenic mouse was generated with cardiomyocyte-specific expression of an ion pore-disruptive Orai1R91W mutant (C-dnO1). Synthetic chemistry and channel screening strategies were used to develop 4-(2,5-dimethoxyphenyl)-N-[(pyridin-4-yl)methyl]aniline (hereafter referred to as JPIII), a small-molecule Orai1 channel inhibitor suitable for in vivo delivery. RESULTS: Adult mice subjected to transverse aortic constriction (TAC) developed cardiac hypertrophy and reduced ventricular function associated with increased Orai1 expression and Orai1-dependent SOCE (assessed by Mn2+ influx). C-dnO1 mice displayed normal cardiac electromechanical function and cellular excitation-contraction coupling despite reduced Orai1-dependent SOCE. Five weeks after TAC, C-dnO1 mice were protected from systolic dysfunction (assessed by preserved left ventricular fractional shortening and ejection fraction) even if increased cardiac mass and prohypertrophic markers induction were observed. This is correlated with a protection from TAC-induced cellular Ca2+ signaling alterations (increased SOCE, decreased [Ca2+]i transients amplitude and decay rate, lower SR Ca2+ load and depressed cellular contractility) and SERCA2a downregulation in ventricular cardiomyocytes from C-dnO1 mice, associated with blunted Pyk2 signaling. There was also less fibrosis in heart sections from C-dnO1 mice after TAC. Moreover, 3 weeks treatment with JPIII following 5 weeks of TAC confirmed the translational relevance of an Orai1 inhibition strategy during hypertrophic insult. CONCLUSIONS: The findings suggest a key role of cardiac Orai1 channels and the potential for Orai1 channel inhibitors as inotropic therapies for maintaining contractility reserve after hypertrophic stress.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/metabolism , Ventricular Function, Left , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , ORAI1 Protein/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
4.
Int J Mol Sci ; 22(19)2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34639190

ABSTRACT

Calcium signaling plays a vital role in the regulation of various cellular processes, including activation, proliferation, and differentiation of T-lymphocytes, which is mediated by ORAI1 and potassium (K+) channels. These channels have also been identified as highly attractive therapeutic targets for immune-related diseases. Licochalcone A is a licorice-derived chalconoid known for its multifaceted beneficial effects in pharmacological treatments, including its anti-inflammatory, anti-asthmatic, antioxidant, antimicrobial, and antitumorigenic properties. However, its anti-inflammatory effects involving ion channels in lymphocytes remain unclear. Thus, the present study aimed to investigate whether licochalcone A inhibits ORAI1 and K+ channels in T-lymphocytes. Our results indicated that licochalcone A suppressed all three channels (ORAI1, Kv1.3, and KCa3.1) in a concentration-dependent matter, with IC50 values of 2.97 ± 1.217 µM, 0.83 ± 1.222 µM, and 11.21 ± 1.07 µM, respectively. Of note, licochalcone A exerted its suppressive effects on the IL-2 secretion and proliferation in CD3 and CD28 antibody-induced T-cells. These results indicate that the use of licochalcone A may provide an effective treatment strategy for inflammation-related immune diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chalcones/pharmacology , Gene Expression Regulation/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Kv1.3 Potassium Channel/antagonists & inhibitors , ORAI1 Protein/antagonists & inhibitors , T-Lymphocytes/drug effects , Calcium/metabolism , Calcium Signaling , HEK293 Cells , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Jurkat Cells , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
Biochem Biophys Res Commun ; 523(1): 153-158, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31839216

ABSTRACT

Brain capillary endothelial cells (BCECs) form the blood-brain barrier (BBB) and play an essential role in the regulation of its functions. Oxidative stress accumulates excessive reactive oxygen species (ROS) and facilitates the death of BCECs, leading to a dysfunctional BBB. However, the mechanisms underlying the death of BCECs under oxidative stress remain unclear. In the present study, the effects of oxidative stress on cell viability, ROS production, intracellular Ca2+ concentration, and protein expression were examined using a cell line derived from bovine BCECs, t-BBEC117. When t-BBEC117 cells were exposed to oxidative stress induced by hydrogen peroxide (H2O2, 10-100 µM), cell growth was inhibited in a dose-dependent manner. Oxidative stress by 30 µM H2O2 increased the production of ROS and its effects were blocked by the ROS scavenger, 10 mM N-acetyl-l-cysteine (NAC). In addition, oxidative stress reduced store-operated Ca2+ entry (SOCE) and this decrease was recovered by NAC or the Orai channel activator, 5 µM 2-aminoethyl diphenylborinate (2-APB). The siRNA knockdown of Orai1 revealed that Orai1 was mainly responsible for SOCE channels and its activity was decreased by oxidative stress. However, the protein expression of Orai1 and STIM1 was not affected by oxidative stress. Oxidative stress-induced cell death was rescued by 2-APB, NAC, or the STIM-Orai activating region. In conclusion, oxidative stress reduces Orai1-mediated SOCE and, thus, facilitates the death of BCECs.


Subject(s)
Acetylcysteine/pharmacology , Calcium/metabolism , Endothelial Cells/drug effects , ORAI1 Protein/antagonists & inhibitors , Oxidative Stress , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cattle , Cell Death/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , ORAI1 Protein/metabolism , Oxidative Stress/drug effects
6.
Int J Mol Sci ; 21(11)2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32521790

ABSTRACT

This study was undertaken to test two therapies for acute kidney injury (AKI) prevention, IGF-1, which is renal protective, and BTP-2, which is a calcium entry (SOCE) inhibitor. We utilized lipopolysaccharide (LPS) IP, as a systemic model of AKI and studied in five groups of animals. Three experiments showed that at 7 days: (1) LPS significantly reduced serum IGF-1 and intramuscular IGF-I in vivo gene therapy rescued this deficiency. (2) Next, at the 7-day time point, our combination therapy,compared to the untreated group,caused a significant increase in survival, which was noteworthy because all of the untreated animals died in 72 hrs. (3) The four pathways associated with inflammation, including (A) increase in cytosolic calcium, (B) elaboration of proinflammatory cytokines, (C) impairment of vascular integrity, and (D) cell injury, were adversely affected in renal tissue by LPS, using a sublethal dose of LPS. The expression of several genes was measured in each of the above pathways. The combined therapy of IGF-1 and BTP-2 caused a favorable gene expression response in all four pathways. Our current study was an AKI study, but these pathways are also involved in other types of severe inflammation, including sepsis, acute respiratory distress syndrome, and probably severe coronavirus infection.


Subject(s)
Acute Kidney Injury/pathology , Insulin-Like Growth Factor I/genetics , Acute Kidney Injury/mortality , Acute Kidney Injury/therapy , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Cytokines/genetics , Cytokines/metabolism , Cytoplasm/metabolism , Disease Models, Animal , Female , Gene Expression/drug effects , Genetic Therapy , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/deficiency , Kidney/metabolism , Kidney/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/metabolism , Survival Rate
7.
J Physiol ; 597(2): 561-582, 2019 01.
Article in English | MEDLINE | ID: mdl-30382595

ABSTRACT

KEY POINTS: Gain-of-function mutations in the highly selective Ca2+ channel ORAI1 cause tubular aggregate myopathy (TAM) characterized by muscular pain, weakness and cramping. TAM-associated mutations in ORAI1 first and third transmembrane domain facilitate channel opening by STIM1, causing constitutive Ca2+ influx and increasing the currents evoked by Ca2+ store depletion. Mutation V107M additionally decreases the channel selectivity for Ca2+ ions and its inhibition by acidic pH, while mutation T184M does not alter the channel sensitivity to pH or to reactive oxygen species. The ORAI blocker GSK-7975A prevents the constitutive activity of TAM-associated channels and might be used in therapy for patients suffering from TAM. ABSTRACT: Skeletal muscle differentiation relies on store-operated Ca2+ entry (SOCE) mediated by STIM proteins linking the depletion of endoplasmic/sarcoplasmic reticulum Ca2+ stores to the activation of membrane Ca2+ -permeable ORAI channels. Gain-of-function mutations in STIM1 or ORAI1 isoforms cause tubular aggregate myopathy (TAM), a skeletal muscle disorder with muscular pain, weakness and cramping. Here, we characterize two overactive ORAI1 mutants from patients with TAM: V107M and T184M, located in the first and third transmembrane domain of the channel. When ectopically expressed in HEK-293T cells or human primary myoblasts, the mutated channels increased basal and store-operated Ca2+ entry. The constitutive activity of V107M, L138F, T184M and P245L mutants was prevented by low concentrations of GSK-7975A while the G98S mutant was resistant to inhibition. Electrophysiological recordings confirmed ORAI1-V107M constitutive activity and revealed larger STIM1-gated V107M- and T184M-mediated currents with conserved fast and slow Ca2+ -dependent inactivation. Mutation V107M altered the channel selectivity for Ca2+ ions and conferred resistance to acidic inhibition. Ca2+ imaging and molecular dynamics simulations showed a preserved sensitivity of T184M to the negative regulation by reactive oxygen species. Both mutants were able to mediate SOCE in Stim1-/- /Stim2-/- mouse embryonic fibroblasts expressing the binding-deficient STIM1-F394H mutant, indicating a higher sensitivity for STIM1-mediated gating, with ORAI1-T184M gain-of-function being strictly dependent on STIM1. These findings provide new insights into the permeation and regulatory properties of ORAI1 mutants that might translate into therapies against diseases with gain-of-function mutations in ORAI1.


Subject(s)
Ion Channel Gating , Myopathies, Structural, Congenital/genetics , ORAI1 Protein/genetics , Animals , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling , Fibroblasts/physiology , Gain of Function Mutation , HEK293 Cells , Humans , Mice, Knockout , Myoblasts/physiology , Myopathies, Structural, Congenital/physiopathology , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/chemistry , ORAI1 Protein/physiology , Protein Domains , Pyrazoles/pharmacology , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 2/genetics
8.
J Physiol ; 597(12): 3085-3105, 2019 06.
Article in English | MEDLINE | ID: mdl-31050811

ABSTRACT

KEY POINTS: This work confirms previous reports that CM4620, a small molecule inhibitor of Ca2+ entry via store operated Ca2+ entry (SOCE) channels formed by stromal interaction molecule 1 (STIM1)/Orai complexes, attenuates acinar cell pathology and acute pancreatitis in mouse experimental models. Here we report that intravenous administration of CM4620 reduces the severity of acute pancreatitis in the rat, a hitherto untested species. Using CM4620, we probe further the mechanisms whereby SOCE via STIM1/Orai complexes contributes to the disease in pancreatic acinar cells, supporting a role for endoplasmic reticulum stress/cell death pathways in these cells. Using CM4620, we show that SOCE via STIM1/Orai complexes promotes neutrophil oxidative burst and inflammatory gene expression during acute pancreatitis, including in immune cells which may be either circulating or invading the pancreas. Using CM4620, we show that SOCE via STIM1/Orai complexes promotes activation and fibroinflammatory gene expression within pancreatic stellate cells. ABSTRACT: Key features of acute pancreatitis include excess cellular Ca2+ entry driven by Ca2+ depletion from the endoplasmic reticulum (ER) and subsequent activation of store-operated Ca2+ entry (SOCE) channels in the plasma membrane. In several cell types, including pancreatic acinar, stellate cells (PaSCs) and immune cells, SOCE is mediated via channels composed primarily of Orai1 and stromal interaction molecule 1 (STIM1). CM4620, a selective Orai1 inhibitor, prevents Ca2+ entry in acinar cells. This study investigates the effects of CM4620 in preventing or reducing acute pancreatitis features and severity. We tested the effects of CM4620 on SOCE, trypsinogen activation, acinar cell death, activation of NFAT and NF-κB, and inflammatory responses in ex vivo and in vivo rodent models of acute pancreatitis and human pancreatic acini. We also examined whether CM4620 inhibited cytokine release in immune cells, fibro-inflammatory responses in PaSCs, and oxidative burst in neutrophils, all cell types participating in pancreatitis. CM4620 administration to rats by i.v. infusion starting 30 min after induction of pancreatitis significantly diminished pancreatitis features including pancreatic oedema, acinar cell vacuolization, intrapancreatic trypsin activity, cell death signalling and acinar cell death. CM4620 also decreased myeloperoxidase activity and inflammatory cytokine expression in pancreas and lung tissues, fMLF peptide-induced oxidative burst in human neutrophils, and cytokine production in human peripheral blood mononuclear cells (PBMCs) and rodent PaSCs, indicating that Orai1/STIM1 channels participate in the inflammatory responses of these cell types during acute pancreatitis. These findings support pathological Ca2+ entry-mediated cell death and proinflammatory signalling as central mechanisms in acute pancreatitis pathobiology.


Subject(s)
Amidines/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Calcium Channel Blockers/therapeutic use , ORAI1 Protein/antagonists & inhibitors , Pancreatitis/drug therapy , Proline/analogs & derivatives , Acinar Cells/metabolism , Amidines/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Ceruletide , Cytokines/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Male , Mice, Inbred C57BL , Pancreatic Stellate Cells/metabolism , Pancreatitis/chemically induced , Pancreatitis/immunology , Pancreatitis/metabolism , Peroxidase/metabolism , Proline/pharmacology , Proline/therapeutic use , Rats , Superoxides/metabolism
9.
FASEB J ; 32(1): 404-416, 2018 01.
Article in English | MEDLINE | ID: mdl-28928244

ABSTRACT

Zinc, an essential micronutrient, has a cancer preventive role. Zinc deficiency has been shown to contribute to the progression of esophageal cancer. Orai1, a store-operated Ca2+ entry (SOCE) channel, was previously reported to be highly expressed in tumor tissues removed from patients with esophageal squamous cell carcinoma (ESCC) with poor prognosis, and elevation of its expression contributes to both hyperactive intracellular Ca2+ oscillations and fast cell proliferation in human ESCC cells. However, the molecular basis of cancer preventive functions of zinc and its association with Orai1-mediated cell proliferation remains unknown. The present study shows that zinc supplementation significantly inhibits proliferation of ESCC cell lines and that the effect of zinc is reversible with N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine, a specific Zn2+ chelator, whereas nontumorigenic esophageal epithelial cells are significantly less sensitive to zinc treatment. Fluorescence live cell imaging revealed that extracellular Zn2+ exerted rapid inhibitory effects on Orai1-mediated SOCE and on intracellular Ca2+ oscillations in the ESCC cells. Knockdown of Orai1 or expression of Orai1 mutants with compromised zinc binding significantly diminished sensitivity of the cancer cells to zinc treatment in both SOCE and cell proliferation analyses. These data suggest that zinc may inhibit cell proliferation of esophageal cancer cells through Orai1-mediated intracellular Ca2+ oscillations and reveal a possible molecular basis for zinc-induced cancer prevention and Orai1-SOCE signaling pathway in cancer cells.-Choi, S., Cui, C., Luo, Y., Kim, S.-H., Ko, J.-K., Huo, X., Ma, J., Fu, L.-W., Souza, R. F., Korichneva, I., Pan, Z. Selective inhibitory effects of zinc on cell proliferation in esophageal squamous cell carcinoma through Orai1.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , ORAI1 Protein/metabolism , Zinc/pharmacology , Amino Acid Substitution , Calcium Signaling/drug effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Ethylenediamines/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Models, Biological , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics
10.
J Cell Mol Med ; 22(10): 4688-4699, 2018 10.
Article in English | MEDLINE | ID: mdl-30117680

ABSTRACT

Our recent study showed that bradykinin increases cell cycling progression and migration of human cardiac c-Kit+ progenitor cells by activating pAkt and pERK1/2 signals. This study investigated whether bradykinin-mediated Ca2+ signalling participates in regulating cellular functions in cultured human cardiac c-Kit+ progenitor cells using laser scanning confocal microscopy and biochemical approaches. It was found that bradykinin increased cytosolic free Ca2+ ( Cai2+ ) by triggering a transient Ca2+ release from ER IP3Rs followed by sustained Ca2+ influx through store-operated Ca2+ entry (SOCE) channel. Blockade of B2 receptor with HOE140 or IP3Rs with araguspongin B or silencing IP3R3 with siRNA abolished both Ca2+ release and Ca2+ influx. It is interesting to note that the bradykinin-induced cell cycle progression and migration were not observed in cells with siRNA-silenced IP3R3 or the SOCE component TRPC1, Orai1 or STIM1. Also the bradykinin-induced increase in pAkt and pERK1/2 as well as cyclin D1 was reduced in these cells. These results demonstrate for the first time that bradykinin-mediated increase in free Cai2+ via ER-IP3R3 Ca2+ release followed by Ca2+ influx through SOCE channel plays a crucial role in regulating cell growth and migration via activating pAkt, pERK1/2 and cyclin D1 in human cardiac c-Kit+ progenitor cells.


Subject(s)
Bradykinin/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Proto-Oncogene Proteins c-kit/genetics , Stem Cells/drug effects , Cations, Divalent , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Gene Expression Regulation , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ion Transport/drug effects , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/cytology , Myocardium/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Quinolizines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Stromal Interaction Molecule 1/antagonists & inhibitors , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
11.
J Biol Chem ; 292(20): 8158-8173, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28325835

ABSTRACT

The concentration of free cytosolic Ca2+ and the voltage across the plasma membrane are major determinants of cell function. Ca2+-permeable non-selective cationic channels are known to regulate these parameters, but understanding of these channels remains inadequate. Here we focus on transient receptor potential canonical 4 and 5 proteins (TRPC4 and TRPC5), which assemble as homomers or heteromerize with TRPC1 to form Ca2+-permeable non-selective cationic channels in many mammalian cell types. Multiple roles have been suggested, including in epilepsy, innate fear, pain, and cardiac remodeling, but limitations in tools to probe these channels have restricted progress. A key question is whether we can overcome these limitations and develop tools that are high-quality, reliable, easy to use, and readily accessible for all investigators. Here, through chemical synthesis and studies of native and overexpressed channels by Ca2+ and patch-clamp assays, we describe compound 31, a remarkable small-molecule inhibitor of TRPC1/4/5 channels. Its potency ranged from 9 to 1300 pm, depending on the TRPC1/4/5 subtype and activation mechanism. Other channel types investigated were unaffected, including TRPC3, TRPC6, TRPV1, TRPV4, TRPA1, TRPM2, TRPM8, and store-operated Ca2+ entry mediated by Orai1. These findings suggest identification of an important experimental tool compound, which has much higher potency for inhibiting TRPC1/4/5 channels than previously reported agents, impressive specificity, and graded subtype selectivity within the TRPC1/4/5 channel family. The compound should greatly facilitate future studies of these ion channels. We suggest naming this TRPC1/4/5-inhibitory compound Pico145.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , TRPC Cation Channels/antagonists & inhibitors , Calcium/metabolism , HEK293 Cells , Humans , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
12.
Am J Physiol Heart Circ Physiol ; 314(2): H359-H369, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29101179

ABSTRACT

Endothelial dysfunction in chronic hypoxia (CH)-induced pulmonary hypertension is characterized by reduced store-operated Ca2+ entry (SOCE) and diminished Ca2+-dependent production of endothelium-derived vasodilators. We recently reported that SOCE in pulmonary arterial endothelial cells (PAECs) is tightly regulated by membrane cholesterol and that decreased membrane cholesterol is responsible for impaired SOCE after CH. However, the ion channels involved in cholesterol-sensitive SOCE are unknown. We hypothesized that cholesterol facilitates SOCE in PAECs through the interaction of Orai1 and stromal interaction molecule 1 (STIM1). The role of cholesterol in Orai1-mediated SOCE was initially assessed using CH exposure in rats (4 wk, 380 mmHg) as a physiological stimulus to decrease PAEC cholesterol. The effects of Orai1 inhibition with AnCoA4 on SOCE were examined in isolated PAEC sheets from control and CH rats after cholesterol supplementation, substitution of endogenous cholesterol with epicholesterol (Epichol), or vehicle treatment. Whereas cholesterol restored endothelial SOCE in CH rats, both Epichol and AnCoA4 attenuated SOCE only in normoxic controls. The Orai1 inhibitor had no further effect in cells pretreated with Epichol. Using cultured pulmonary endothelial cells to allow better mechanistic analysis of the molecular components of cholesterol-regulated SOCE, we found that Epichol, AnCoA4, and Orai1 siRNA each inhibited SOCE compared with their respective controls. Epichol had no additional effect after knockdown of Orai1. Furthermore, Epichol substitution significantly reduced STIM1-Orai1 interactions as assessed by a proximity ligation assay. We conclude that membrane cholesterol is required for the STIM1-Orai1 interaction necessary to elicit endothelial SOCE. Furthermore, reduced PAEC membrane cholesterol after CH limits Orai1-mediated SOCE. NEW & NOTEWORTHY This research demonstrates a novel contribution of cholesterol to regulate the interaction of Orai1 and stromal interaction molecule 1 required for pulmonary endothelial store-operated Ca2+ entry. The results provide a mechanistic basis for impaired pulmonary endothelial Ca2+ influx after chronic hypoxia that may contribute to pulmonary hypertension.


Subject(s)
Calcium Signaling , Cell Membrane/metabolism , Cholesterol/metabolism , Endothelial Cells/metabolism , Hypoxia/metabolism , ORAI1 Protein/metabolism , Pulmonary Artery/metabolism , Animals , Arterial Pressure , Benzodioxoles/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cells, Cultured , Chromones/pharmacology , Chronic Disease , Disease Models, Animal , Down-Regulation , Endothelial Cells/drug effects , Hypoxia/physiopathology , Male , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Stromal Interaction Molecule 1/metabolism
13.
Cell Physiol Biochem ; 48(6): 2273-2285, 2018.
Article in English | MEDLINE | ID: mdl-30114708

ABSTRACT

BACKGROUND/AIMS: Multiple myeloma (MM) is a plasma cell neoplasm which constitutes about 10% of all hematologic malignancies. Despite the development and application of novel agents, MM still undergoes an aggressive and incurable course in the vast majority of patients. Ca2+ is one of the critical regulators of cell migration. Ca2+ influx is essential for the migration of various types of cells including tumor cells. However, the role of store-operated calcium entry (SOC) channels, the only Ca2+ channels of non-excitable cells, has not yet been reported in MM cell survival. METHODS: We evaluated the expression of Stim1 and Orai1 (two key regulators of SOC) in MM tissues and cell lines by immunohistochemical assay, quantitative real-time PCR assay and western blot. MM cell lines were pretreated with pharmacological blockers and siRNAs, and then MM cell proliferation, cell cycle arrest, and apoptosis were examined by FACS (flow cytometry) assay, and Annexin V-FITC/PI staining. The correlation between the expression of Stim1 (or Orai1) level and outcome in MM were assessed by using Progress Free Survival (PFS). RESULTS: Stim1 and Orai1 were both abundantly expressed in MM tissue and MM cell lines. Inhibition of SOCE reduced MM cell viability, and induced cell cycle arrest and apoptosis. Stim1 or Orai1 silencing also reduced cell viability, caused cell apoptosis and cell cycle arrest in MM cell lines. Over-expression of Stim1/Orai1 in MM patients was closely associated with the clinical outcome of MM. CONCLUSION: The Stim1/Orai1-mediated signaling participates in the pathogenesis of MM, which represents an attractive target for future therapeutic intervention.


Subject(s)
Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Adult , Aged , Apoptosis/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Imidazoles/pharmacology , Male , Middle Aged , Multiple Myeloma/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Stromal Interaction Molecule 1/antagonists & inhibitors , Stromal Interaction Molecule 1/genetics
14.
Bioorg Med Chem ; 26(12): 3406-3413, 2018 07 23.
Article in English | MEDLINE | ID: mdl-29776832

ABSTRACT

The proteins Orai1 and STIM1 control store-operated Ca2+ entry (SOCE) into cells. SOCE is important for migration, invasion and metastasis of MDA-MB-231 human triple negative breast cancer (TNBC) cells and has been proposed as a target for cancer drug discovery. Two hit compounds from a medium throughput screen, displayed encouraging inhibition of SOCE in MDA-MB-231 cells, as measured by a Fluorescence Imaging Plate Reader (FLIPR) Ca2+ assay. Following NMR spectroscopic analysis of these hits and reassignment of their structures as 5-hydroxy-5-trifluoromethylpyrazolines, a series of analogues was prepared via thermal condensation reactions between substituted acylhydrazones and trifluoromethyl 1,3-dicarbonyl arenes. Structure-activity relationship (SAR) studies showed that small lipophilic substituents at the 2- and 3-positions of the RHS and 2-, 3- and 4-postions of the LHS terminal benzene rings improved activity, resulting in a novel class of potent and selective inhibitors of SOCE.


Subject(s)
Calcium Channel Blockers/chemistry , ORAI1 Protein/antagonists & inhibitors , Pyrazoles/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium/metabolism , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Cell Line, Tumor , Female , Humans , ORAI1 Protein/metabolism , Protein Array Analysis , Pyrazoles/metabolism , Pyrazoles/pharmacology , Spectrometry, Fluorescence , Structure-Activity Relationship
15.
Am J Physiol Renal Physiol ; 312(6): F1090-F1100, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28298362

ABSTRACT

Collagen IV (Col IV) is a major component of expanded glomerular extracellular matrix in diabetic nephropathy and Smad1 is a key molecule regulating Col IV expression in mesangial cells (MCs). The present study was conducted to determine if Smad1 pathway and Col IV protein abundance were regulated by store-operated Ca2+ entry (SOCE). In cultured human MCs, pharmacological inhibition of SOCE significantly increased the total amount of Smad1 protein. Activation of SOCE blunted high-glucose-increased Smad1 protein content. Treatment of human MCs with ANG II at 1 µM for 15 min, high glucose for 3 days, or TGF-ß1 at 5 ng/ml for 30 min increased the level of phosphorylated Smad1. However, the phosphorylation of Smad1 by those stimuli was significantly attenuated by activation of SOCE. Knocking down Smad1 reduced, but expressing Smad1 increased, the amount of Col IV protein. Furthermore, activation of SOCE significantly attenuated high-glucose-induced Col IV protein production, and blockade of SOCE substantially increased the abundance of Col IV. To further verify those in vitro findings, we downregulated SOCE specifically in MCs in mice using small-interfering RNA (siRNA) against Orai1 (the channel protein mediating SOCE) delivered by the targeted nanoparticle delivery system. Immunohistochemical examinations showed that expression of both Smad1 and Col IV proteins was significantly greater in the glomeruli with positively transfected Orai1 siRNA compared with the glomeruli from the mice without Orai1 siRNA treatment. Taken together, our results indicate that SOCE negatively regulates the Smad1 signaling pathway and inhibits Col IV protein production in MCs.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Signaling , Collagen Type IV/metabolism , Mesangial Cells/metabolism , ORAI1 Protein/metabolism , Smad1 Protein/metabolism , Angiotensin II/metabolism , Animals , Calcium Signaling/drug effects , Cells, Cultured , Gene Expression Regulation , Glucose/pharmacology , Humans , Mesangial Cells/drug effects , Mice , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , Phosphorylation , RNA Interference , Smad1 Protein/genetics , Time Factors , Transfection , Transforming Growth Factor beta1/pharmacology
16.
Am J Physiol Renal Physiol ; 313(3): F729-F739, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28637791

ABSTRACT

Our previous study demonstrated that the abundance of extracellular matrix proteins was suppressed by store-operated Ca2+ entry (SOCE) in mesangial cells (MCs). The present study was conducted to investigate the underlying mechanism focused on the transforming growth factor-ß1 (TGF-ß1)/Smad3 pathway, a critical pathway for ECM expansion in diabetic kidneys. We hypothesized that SOCE suppressed ECM protein expression by inhibiting this pathway in MCs. In cultured human MCs, we observed that TGF-ß1 (5 ng/ml for 15 h) significantly increased Smad3 phosphorylation, as evaluated by immunoblot. However, this response was markedly inhibited by thapsigargin (1 µM), a classical activator of store-operated Ca2+ channels. Consistently, both immunocytochemistry and immunoblot showed that TGF-ß1 significantly increased nuclear translocation of Smad3, which was prevented by pretreatment with thapsigargin. Importantly, the thapsigargin effect was reversed by lanthanum (La3+; 5 µM) and GSK-7975A (10 µM), both of which are selective blockers of store-operated Ca2+ channels. Furthermore, knockdown of Orai1, the pore-forming subunit of the store-operated Ca2+ channels, significantly augmented TGF-ß1-induced Smad3 phosphorylation. Overexpression of Orai1 augmented the inhibitory effect of thapsigargin on TGF-ß1-induced phosphorylation of Smad3. In agreement with the data from cultured MCs, in vivo knockdown of Orai1 specific to MCs using a targeted nanoparticle small interfering RNA delivery system resulted in a marked increase in abundance of phosphorylated Smad3 and in nuclear translocation of Smad3 in the glomerulus of mice. Taken together, our results indicate that SOCE in MCs negatively regulates the TGF-ß1/Smad3 signaling pathway.


Subject(s)
Calcium Signaling , Mesangial Cells/drug effects , ORAI1 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/pharmacology , Active Transport, Cell Nucleus , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Collagen Type IV/metabolism , Enzyme Inhibitors/pharmacology , Fibronectins/metabolism , Humans , Male , Mesangial Cells/metabolism , Mice, Inbred C57BL , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , Phosphorylation , RNA Interference , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Time Factors , Transfection
17.
Cell Physiol Biochem ; 42(4): 1366-1376, 2017.
Article in English | MEDLINE | ID: mdl-28704809

ABSTRACT

BACKGROUND/AIMS: Istaroxime is a validated inotropic Na+/K+ ATPase inhibitor currently in development for the treatment of various cardiac conditions. Recent findings established that this steroidal drug exhibits potent apoptotic responses in prostate tumors in vitro and in vivo, by affecting key signaling orchestrating proliferation and apoptosis, such as c-Myc and caspase 3, Rho GTPases and actin cytoskeleton dynamics. In the present study we examined whether istaroxime is affecting cell motility and analyzed the underlying mechanism in prostate tumor cells. METHODS: Migration was assessed by transwell and wound healing assays, Orai1 and Stim1 abundance by RT-PCR and confocal immunofluorescence microscopy, Fura-2 fluorescence was utilized to determine intracellular Ca2+ and Western blotting for FAK/pFAK measurements. RESULTS: We observed strong inhibition of cell migration in istaroxime treated DU-145 prostate cancer cells. Istaroxime further decreased Orai1 and Stim1 transcript levels and downregulated Orai1 protein expression. Moreover, SOCE was significantly decreased upon istaroxime treatment. Furthermore, istaroxime strikingly diminished phosphorylated FAK levels. Interestingly, the efficacy of istaroxime on the inhibition of DU-145 cell migration was further enhanced by blocking Orai1 with 2-APB and FAK with the specific inhibitor PF-00562271. These results provide strong evidence that istaroxime prevents cell migration and motility of DU-145 prostate tumor cells, an effect at least partially attributed to Orai1 downregulation and FAK de-activation. CONCLUSION: Collectively our results indicate that this enzyme inhibitor, besides its pro-apoptotic action, affects motility of cancer cells, supporting its potential role as a strong candidate for further clinical cancer drug development.


Subject(s)
Cell Movement/drug effects , Epithelial Cells/drug effects , Etiocholanolone/analogs & derivatives , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , ORAI1 Protein/genetics , Sodium Channel Blockers/pharmacology , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Etiocholanolone/pharmacology , Fluorescent Dyes/chemistry , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Fura-2/chemistry , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/metabolism , Phosphorylation/drug effects , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Stromal Interaction Molecule 1/antagonists & inhibitors , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Sulfonamides/pharmacology
18.
Bioorg Med Chem ; 25(1): 440-449, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27856238

ABSTRACT

The Orai1 Ca2+ permeable ion channel is an important component of store operated Ca2+ entry (SOCE) in cells. It's over-expression in basal molecular subtype breast cancers has been linked with poor prognosis, making it a potential target for drug development. We pharmacologically characterised a number of reported inhibitors of SOCE in MDA-MB-231 breast cancer cells using a convenient Fluorescence Imaging Plate Reader (FLIPR) assay, and show that the rank order of their potencies in this assay is the same as those reported in a wide range of published assays. The assay was also used in a screening project seeking novel inhibitors. Following a broad literature survey of classes of calcium channel inhibitors we used simplified ligand structures to query the ZINC on-line database, and following two iterations of refinement selected a novel Orai1-selective dichlorophenyltriazole hit compound. Analogues of this were synthesized and evaluated in the FLIPR assay to develop structure-activity relationships (SAR) for the three domains of the hit; triazole (head), dichlorophenyl (body) and substituted phenyl (tail). For this series, the results suggested the need for a lipophilic tail domain and an out-of-plane twist between the body and tail domains.


Subject(s)
Calcium Channel Blockers/pharmacology , ORAI1 Protein/antagonists & inhibitors , Calcium Channel Blockers/chemical synthesis , Cell Line, Tumor , Databases, Chemical , Drug Stability , Fluorescence , HEK293 Cells , High-Throughput Screening Assays , Humans , Stereoisomerism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacology
19.
J Am Soc Nephrol ; 27(10): 3063-3078, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26940090

ABSTRACT

Evidence supports an important role of Ca2+ release-activated Ca2+ channel protein 1 (Orai1)-mediated Ca2+ entry in the development of renal fibrosis, a common pathologic feature of CKDs that lead to ESRD, but the molecular mechanisms remain unclear. We determined the role of Orai1 calcium channel in renal fibrosis induced by high-fat diet and by unilateral ureteral obstruction. Mouse kidneys with fibrosis had higher levels of Orai1 protein expression than did kidneys without fibrosis. In vivo knockdown of Orai1 with adenovirus harboring Orai1-short hairpin RNA or inhibition of Orai1 with SKF96365 dramatically prevented renal fibrosis and significantly decreased protein expression of fibronectin, α­smooth muscle actin, and TGF­ß1 in the kidney cortex of ApoE-/- mice on a high-fat diet and in the obstructed kidneys of mice with unilateral ureteral obstruction. Compared with kidney biopsy specimens of patients with glomerular minimal change disease, those of patients with fibrotic nephropathy had higher expression levels of Orai1. In cultured human proximal tubule epithelial cells (HK2), knockdown of Orai1 Ca2+ channel with adenovirus-Orai1-short hairpin RNA markedly inhibited TGF-ß1-induced intracellular Ca2+ influx and phosphorylation of smad2/3. Knockdown or blockade of the Orai1 Ca2+ channel in HK2 cells also prevented epithelial-to-mesenchymal transition induced by TGF­ß1. In conclusion, blockade of the Orai1 Ca2+ channel prevented progression of renal fibrosis in mice, likely by suppressing smad2/3 phosphorylation and TGF-ß1-induced epithelial-to-mesenchymal transition. These results render the Orai1 Ca2+ channel a potential therapeutic target against renal fibrosis.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium/metabolism , Imidazoles/pharmacology , Kidney/pathology , ORAI1 Protein/antagonists & inhibitors , Animals , Cells, Cultured , Fibrosis/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Pharm Biol ; 55(1): 1324-1329, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28290212

ABSTRACT

CONTEXT: Spirodela polyrhiza (L.) Schleid. (Lemnaceae), Spirodelae Herba (SH), has been known to relieve inflammation, urticaria and skin symptoms including pruritus, eczema and rash. OBJECTIVE: The effects of SH extract on two calcium ion channels, Orai1 and TRPV3, and their potential as novel therapeutics for atopic dermatitis (AD) were investigated. The regulatory role of Orai1 on mast cell degranulation was evaluated. MATERIALS AND METHODS: The dried leaves of SH were extracted by 70% methanol. Effects of SH extract (100 µg/mL) in an HEK293T cell line overexpressing human Orai1 or TRPV3 were assessed. Ion channel modulation in transfected HEK293T cells was measured using a conventional whole-cell patch-clamp technique. IgE-antigen complex-stimulated mast cell degranulation was measured by ß-hexosaminidase assay with morphological observation after treatment with 20, 50 and 100 µg/mL SH extract. RESULTS: SH extract (100 µg/mL) significantly inhibited Orai1 activity (63.8 ± 0.97%) in Orai1-STIM1 co-overexpressed HEK293T cells. SH extract significantly increased TRPV3 activity (81.29 ± 0.05% at -100 mV) compared with the positive control 2-APB (100 µM), which induced full activation. SH extract inhibited degranulation in IgE-antigen complex-stimulated RBL-2H3 mast cells by decreasing ß-hexosaminidase activity (3.14 ± 0.03, 2.56 ± 0.12 and 2.29 ± 0.08 mU/mg, respectively). CONCLUSION: Our results suggested that SH extract could treat abnormal skin barrier pathologies in AD through modulation of the activities of the calcium ion channels Orai1 and TRPV3 and inhibition of mast cell degranulation. This is the first report of an herbal effect on the modulation of ion channels associated with skin barrier disruption in AD pathogenesis.


Subject(s)
Araceae , Cell Degranulation/drug effects , Mast Cells/drug effects , ORAI1 Protein/antagonists & inhibitors , Plant Extracts/pharmacology , TRPV Cation Channels/agonists , Cell Degranulation/physiology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Mast Cells/physiology , ORAI1 Protein/biosynthesis , Patch-Clamp Techniques/methods , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , TRPV Cation Channels/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL