1
|
Liu X, Pan Z. Store-Operated Calcium Entry in the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:303-333. [DOI: 10.1007/978-981-16-4254-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
2
|
Lopez JJ, Jardin I, Albarrán L, Sanchez-Collado J, Cantonero C, Salido GM, Smani T, Rosado JA. Molecular Basis and Regulation of Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:445-469. [PMID: 31646520 DOI: 10.1007/978-3-030-12457-1_17] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous mechanism for Ca2+ influx in mammalian cells with important physiological implications. Since the discovery of SOCE more than three decades ago, the mechanism that communicates the information about the amount of Ca2+ accumulated in the intracellular Ca2+ stores to the plasma membrane channels and the nature of these channels have been matters of intense investigation and debate. The stromal interaction molecule-1 (STIM1) has been identified as the Ca2+ sensor of the intracellular Ca2+ compartments that activates the store-operated channels. STIM1 regulates two types of store-dependent channels: the Ca2+ release-activated Ca2+ (CRAC) channels, formed by Orai1 subunits, that conduct the highly Ca2+ selective current I CRAC and the cation permeable store-operated Ca2+ (SOC) channels, which consist of Orai1 and TRPC1 proteins and conduct the non-selective current I SOC. While the crystal structure of Drosophila CRAC channel has already been solved, the architecture of the SOC channels still remains unclear. The dynamic interaction of STIM1 with the store-operated channels is modulated by a number of proteins that either support the formation of the functional STIM1-channel complex or protect the cell against Ca2+ overload.
Collapse
Affiliation(s)
- Jose J Lopez
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Isaac Jardin
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain.
| | - Letizia Albarrán
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Jose Sanchez-Collado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Carlos Cantonero
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Gines M Salido
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics and Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/University of Sevilla, Sevilla, Spain
| | - Juan A Rosado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| |
Collapse
|
3
|
Bhuvaneshwari S, Sankaranarayanan K. Structural and Mechanistic Insights of CRAC Channel as a Drug Target in Autoimmune Disorder. Curr Drug Targets 2019; 21:55-75. [PMID: 31556856 DOI: 10.2174/1389450120666190926150258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Calcium (Ca2+) ion is a major intracellular signaling messenger, controlling a diverse array of cellular functions like gene expression, secretion, cell growth, proliferation, and apoptosis. The major mechanism controlling this Ca2+ homeostasis is store-operated Ca2+ release-activated Ca2+ (CRAC) channels. CRAC channels are integral membrane protein majorly constituted via two proteins, the stromal interaction molecule (STIM) and ORAI. Following Ca2+ depletion in the Endoplasmic reticulum (ER) store, STIM1 interacts with ORAI1 and leads to the opening of the CRAC channel gate and consequently allows the influx of Ca2+ ions. A plethora of studies report that aberrant CRAC channel activity due to Loss- or gain-of-function mutations in ORAI1 and STIM1 disturbs this Ca2+ homeostasis and causes several autoimmune disorders. Hence, it clearly indicates that the therapeutic target of CRAC channels provides the space for a new approach to treat autoimmune disorders. OBJECTIVE This review aims to provide the key structural and mechanical insights of STIM1, ORAI1 and other molecular modulators involved in CRAC channel regulation. RESULTS AND CONCLUSION Understanding the structure and function of the protein is the foremost step towards improving the effective target specificity by limiting their potential side effects. Herein, the review mainly focusses on the structural underpinnings of the CRAC channel gating mechanism along with its biophysical properties that would provide the solid foundation to aid the development of novel targeted drugs for an autoimmune disorder. Finally, the immune deficiencies caused due to mutations in CRAC channel and currently used pharmacological blockers with their limitation are briefly summarized.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai -600 044, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai -600 044, India
| |
Collapse
|
4
|
Bhuvaneshwari S, Sankaranarayanan K. Identification of potential CRAC channel inhibitors: Pharmacophore mapping, 3D-QSAR modelling, and molecular docking approach. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2019; 30:81-108. [PMID: 30773908 DOI: 10.1080/1062936x.2019.1566172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Indexed: 06/09/2023]
Abstract
Upregulation of store-operated Ca2+ influx via ORAI1, an integral component of the CRAC channel, is responsible for abnormal cytokine release in active rheumatoid arthritis, and therefore ORAI1 has been proposed as an attractive molecular target. In this study, we attempted to predict the mechanical insights of ORAI1 inhibitors through pharmacophore modelling, 3D-QSAR, molecular docking and free energy analysis. Various hypotheses of pharmacophores were generated and from that, a pharmacophore hypothesis with two hydrogen bond acceptors, one hydrogen bond donor and two aromatic rings (AADRR) resulted in a statistically significant 3D-QSAR model (r2 = 0.84 and q2 = 0.74). We believe that the obtained statistical model is a reliable QSAR model for the diverse dataset of inhibitors against the IL-2 production assay. The visualization of contours in active and inactive compounds generated from the 3D-QSAR models and molecular docking studies revealed major interaction with GLN108, HIS113 and ASP114, and interestingly, these residues are located near the Ca2+ selectivity filter region. Free energy binding analysis revealed that Coulomb energy, van der Waals energy and non-polar solvation terms are more favourable for ligand binding. Thus, the present study provides the physical and chemical requirements for the development of novel ORAI1 inhibitors with improved biological activity.
Collapse
Affiliation(s)
- S Bhuvaneshwari
- a Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University , Chennai , India
| | - K Sankaranarayanan
- a Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University , Chennai , India
| |
Collapse
|
5
|
Lange I, Moschny J, Tamanyan K, Khutsishvili M, Atha D, Borris RP, Koomoa DL. Scrophularia orientalis extract induces calcium signaling and apoptosis in neuroblastoma cells. Int J Oncol 2016; 48:1608-16. [PMID: 26848085 PMCID: PMC4777595 DOI: 10.3892/ijo.2016.3373] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022] Open
Abstract
Effective neuroblastoma (NB) treatments are still limited despite treatment options available today. Therefore, this study attempted to identify novel plant extracts that have anticancer effects. Cytotoxicity and increased intracellular calcium levels were determined using the Sulforhodamine B (SRB) assay and Fluo4-AM (acetoxymethyl) staining and fluorescence microscopy in NB cells in order to screen a library of plant extracts. The current study examined the anticancer effects of a dichloromethane extract from Scrophularia orientalis L. (Scrophulariaceae), a plant that has been used in Traditional Chinese Medicine. This extract contained highly potent agents that significantly reduced cell survival and increased calcium levels in NB cells. Further analysis revealed that cell death induced by this extract was associated with intracellular calcium release, opening of the MPTP, caspase 3- and PARP-cleavage suggesting that this extract induced aberrant calcium signaling that resulted in apoptosis via the mitochondrial pathway. Therefore, agents from Scrophularia orientalis may have the potential to lead to new chemo therapeutic anticancer drugs. Furthermore, targeting intracellular calcium signaling may be a novel strategy to develop more effective treatments for NB.
Collapse
Affiliation(s)
- Ingo Lange
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| | - Julia Moschny
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| | - Kamilla Tamanyan
- Institute of Botany, National Academy of Sciences, Yerevan, Armenia
| | - Manana Khutsishvili
- National Herbarium of Georgia, Institute of Botany, Ilia State University, Tbilisi, Georgia
| | - Daniel Atha
- The New York Botanical Garden, Bronx, NY, USA
| | - Robert P Borris
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Dana-Lynn Koomoa
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| |
Collapse
|
6
|
Migration and Phagocytic Ability of Activated Microglia During Post-natal Development is Mediated by Calcium-Dependent Purinergic Signalling. Mol Neurobiol 2015; 53:944-954. [PMID: 25575683 DOI: 10.1007/s12035-014-9064-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/09/2014] [Indexed: 01/10/2023]
Abstract
Microglia play an important role in synaptic pruning and controlled phagocytosis of neuronal cells during developmental stages. However, the mechanisms that regulate these functions are not completely understood. The present study was designed to investigate the role of purinergic signalling in microglial migration and phagocytic activity during post-natal brain development. One-day-old BALB/c mice received lipopolysaccharide (LPS) and/or a purinergic analogue (2-methylthioladenosine-5'-diphosphate; 2MeSADP), intracerebroventrically (i.c.v.). Combined administration of LPS and 2MeSADP resulted in activation of microglia as evident from increased expression of ionised calcium-binding adapter molecule 1 (Iba1). Activated microglia showed increased expression of purinergic receptors (P2Y2, P2Y6 and P2Y12). LPS either alone or in combination with 2MeSADP induced the expression of Na(+)/Ca(2+) exchanger (NCX-1) and P/Q-type Ca(2+) channels along with MARCKS-related protein (MRP), which is an integral component of cell migration machinery. In addition, LPS and 2MeSADP administration induced the expression of microglial CD11b and DAP12 (DNAX-activation protein 12), which are known to be involved in phagocytosis of neurons during development. Interestingly, administration of thapsigargin (TG), a specific Ca(2+)-ATPase inhibitor of endoplasmic reticulum, prevented the LPS/2MeSADP-induced microglial activation and migration by down-regulating the expression of Iba1 and MRP, respectively. Moreover, TG also reduced the LPS/2MeSADP-induced expression of CD11b/DAP12. Taken together, the findings reveal for the first time that Ca(2+)-mediated purinergic receptors regulate the migration and phagocytic ability of microglia during post-natal brain development.
Collapse
|
7
|
Zhu M, Chen L, Zhao P, Zhou H, Zhang C, Yu S, Lin Y, Yang X. Store-operated Ca(2+) entry regulates glioma cell migration and invasion via modulation of Pyk2 phosphorylation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014. [PMID: 25433371 DOI: 10.1186/preaccept-3101393591453932] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND The ubiquitous second messenger Ca(2+) has been demonstrated to play an important role in cancer progression. Store-operated Ca(2+) entry (SOCE) is the main Ca(2+) entry pathway regulating intracellular Ca(2+) concentration in a variety of cancer types. The present study aimed to explore the specific mechanisms of SOCE in the processes of glioma migration and invasion. METHODS The expression of Orai1, a key component of SOCE, was examined in glioma samples and glioma cell lines by immunohistochemistry and western blot analysis. Both pharmacological intervention and RNA interference were employed to investigate the role of SOCE in glioma cell migration and invasion in vitro. The intracellular Ca(2+) was certified through Fluo-4/AM based Ca(2+) measurement. The effect of SOCE on cell viability, migration, and invasion was explored by methyl thiazolyl tetrazolium (MTT) assay, wound healing assay, transwell invasion assay. Western blot analysis and immunofluorescence assay were used to observe the changes of downstream related protein and cell morpholog. RESULTS Orai1 expression was elevated in glioma tissues and several glioma cell lines compared with non-neoplastic brain tissues. Either inhibition of SOCE by a pharmacological inhibitor or Orai1 downregulation suppressed glioma cell migration and invasion. However, re-expression of Orai1 could rescue glioma cell motility. Furthermore, phosphorylation of proline-rich tyrosine kinase 2 (Pyk2) participated in the mechanisms by which SOCE regulated focal adhesion turnover and epithelial-to-mesenchymal (-like) transition in glioma cells, both of which are considered to be critical for tumor progression. CONCLUSIONS The SOCE-Pyk2 pathway is essential for glioma migration and invasion. The study indicates the potential value of Orai1 as a molecular target for anti-invasion therapy.
Collapse
Affiliation(s)
- Meng Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Lei Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Pengfei Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Hua Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Yu Lin
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| |
Collapse
|
8
|
Zhu M, Chen L, Zhao P, Zhou H, Zhang C, Yu S, Lin Y, Yang X. Store-operated Ca(2+) entry regulates glioma cell migration and invasion via modulation of Pyk2 phosphorylation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:98. [PMID: 25433371 PMCID: PMC4258251 DOI: 10.1186/s13046-014-0098-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/14/2014] [Indexed: 01/08/2023]
Abstract
Background The ubiquitous second messenger Ca2+ has been demonstrated to play an important role in cancer progression. Store-operated Ca2+ entry (SOCE) is the main Ca2+ entry pathway regulating intracellular Ca2+ concentration in a variety of cancer types. The present study aimed to explore the specific mechanisms of SOCE in the processes of glioma migration and invasion. Methods The expression of Orai1, a key component of SOCE, was examined in glioma samples and glioma cell lines by immunohistochemistry and western blot analysis. Both pharmacological intervention and RNA interference were employed to investigate the role of SOCE in glioma cell migration and invasion in vitro. The intracellular Ca2+ was certified through Fluo-4/AM based Ca2+ measurement. The effect of SOCE on cell viability, migration, and invasion was explored by methyl thiazolyl tetrazolium (MTT) assay, wound healing assay, transwell invasion assay. Western blot analysis and immunofluorescence assay were used to observe the changes of downstream related protein and cell morpholog. Results Orai1 expression was elevated in glioma tissues and several glioma cell lines compared with non-neoplastic brain tissues. Either inhibition of SOCE by a pharmacological inhibitor or Orai1 downregulation suppressed glioma cell migration and invasion. However, re-expression of Orai1 could rescue glioma cell motility. Furthermore, phosphorylation of proline-rich tyrosine kinase 2 (Pyk2) participated in the mechanisms by which SOCE regulated focal adhesion turnover and epithelial-to-mesenchymal (−like) transition in glioma cells, both of which are considered to be critical for tumor progression. Conclusions The SOCE-Pyk2 pathway is essential for glioma migration and invasion. The study indicates the potential value of Orai1 as a molecular target for anti-invasion therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13046-014-0098-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meng Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Lei Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Pengfei Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Hua Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Yu Lin
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154# Anshan Road, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| |
Collapse
|
9
|
Collins HE, Zhu-Mauldin X, Marchase RB, Chatham JC. STIM1/Orai1-mediated SOCE: current perspectives and potential roles in cardiac function and pathology. Am J Physiol Heart Circ Physiol 2013; 305:H446-58. [PMID: 23792674 PMCID: PMC3891250 DOI: 10.1152/ajpheart.00104.2013] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Store-operated Ca²⁺ entry (SOCE) is critical for Ca²⁺ signaling in nonexcitable cells; however, its role in the regulation of cardiomyocyte Ca²⁺ homeostasis has only recently been investigated. The increased understanding of the role of stromal interaction molecule 1 (STIM1) in regulating SOCE combined with recent studies demonstrating the presence of STIM1 in cardiomyocytes provides support that this pathway co-exists in the heart with the more widely recognized Ca²⁺ handling pathways associated with excitation-contraction coupling. There is now substantial evidence that STIM1-mediated SOCE plays a key role in mediating cardiomyocyte hypertrophy, both in vitro and in vivo, and there is growing support for the contribution of SOCE to Ca²⁺ overload associated with ischemia/reperfusion injury. Here, we provide an overview of our current understanding of the molecular regulation of SOCE and discuss the evidence supporting the role of STIM1/Orai1-mediated SOCE in regulating cardiomyocyte function.
Collapse
Affiliation(s)
- Helen E Collins
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | |
Collapse
|
10
|
Salerno C, Capuozzo E. Effects of the semisynthetic bis-indole derivative KAR-2 on store-operated calcium entry in human neutrophils. Arch Biochem Biophys 2013; 537:133-7. [PMID: 23876240 DOI: 10.1016/j.abb.2013.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/02/2013] [Accepted: 07/09/2013] [Indexed: 02/02/2023]
Abstract
We studied the effect of KAR-2 on cytosolic Ca(2+) level in human neutrophils by using a fluorescent dye (Fura-2) trapped in the cells. KAR-2 is a semisynthetic bis-indole derivative that shares vinblastine anti-microtubular properties, but does not share the vinblastine antagonistic effect on calmodulin. Therefore KAR-2 offers a convenient mean of studying the effect of microtubule destabilization, without concomitant calmodulin alterations. We found that KAR-2 induces Ca(2+) release from intracellular stores, whereby the stores are depleted. In addition KAR-2 reduces store-operated entry of extracellular Ca(2+) induced by agonists such as thapsigargin or ATP. On the other hand, in Ca(2+) refilled cells, KAR-2 promotes limited entry of extracellular Ca(2+) in the absence of agonist, but still interferes prominently with Ca(2+) entry triggered by ATP and with Ca(2+) uptake by intracellular stores. We suggest that Ca(2+) traffic through the plasma membrane is operated by two diverse pathways: the prominent pathway is interfered with by microtubule destabilization, while an alternate and minor pathway is actually favored (or uncovered) following microtubule destabilization.
Collapse
Affiliation(s)
- Costantino Salerno
- Department of Biochemical Sciences, University of Rome La Sapienza, Italy.
| | | |
Collapse
|
11
|
Pan LJ, Zhang ZC, Zhang ZY, Wang WJ, Xu Y, Zhang ZM. Effects and mechanisms of store-operated calcium channel blockade on hepatic ischemia-reperfusion injury in rats. World J Gastroenterol 2012; 18:356-67. [PMID: 22294842 PMCID: PMC3261531 DOI: 10.3748/wjg.v18.i4.356] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/18/2011] [Accepted: 07/25/2011] [Indexed: 02/06/2023] Open
Abstract
AIM To further investigate the important role of store-operated calcium channels (SOCs) in rat hepatocytes and to explore the effects of SOC blockers on hepatic ischemia-reperfusion injury (HIRI). METHODS Using freshly isolated hepatocytes from a rat model of HIRI (and controls), we measured cytosolic free Ca(2+) concentration (by calcium imaging), net Ca(2+) fluxes (by a non-invasive micro-test technique), the SOC current (I(SOC); by whole-cell patch-clamp recording), and taurocholate secretion [by high-performance liquid chromatography and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays]. RESULTS Ca(2+) oscillations and net Ca(2+) fluxes mediated by Ca(2+) entry via SOCs were observed in rat hepatocytes. I(SOC) was significantly higher in HIRI groups than in controls (57.0 ± 7.5 pA vs 31.6 ± 2.7 pA, P < 0.05) and was inhibited by La(3+). Taurocholate secretion by hepatocytes into culture supernatant was distinctly lower in HIRI hepatocytes than in controls, an effect reversed by SOC blockers. CONCLUSION SOCs are pivotal in HIRI. SOC blockers protected against HIRI and assisted the recovery of secretory function in hepatocytes. Thus, they are likely to become a novel class of effective drugs for prevention or therapy of HIRI patients in the future.
Collapse
Affiliation(s)
- Li-Jie Pan
- Department of General Surgery, Digestive Medical Center, The First Affiliated Hospital, School of Medicine, Tsinghua University, Beijing 100016, China
| | | | | | | | | | | |
Collapse
|
12
|
Giachini FR, Lima VV, Hannan JL, Carneiro FS, Webb RC, Tostes RC. STIM1/Orai1-mediated store-operated Ca2+ entry: the tip of the iceberg. Braz J Med Biol Res 2011; 44:1080-7. [PMID: 22002090 DOI: 10.1590/s0100-879x2011007500133] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 09/05/2011] [Indexed: 11/22/2022] Open
Abstract
Highly efficient mechanisms regulate intracellular calcium (Ca2+) levels. The recent discovery of new components linking intracellular Ca2+ stores to plasma membrane Ca2+ entry channels has brought new insight into the understanding of Ca2+ homeostasis. Stromal interaction molecule 1 (STIM1) was identified as a Ca2+ sensor essential for Ca2+ store depletion-triggered Ca2+ influx. Orai1 was recognized as being an essential component for the Ca2+ release-activated Ca2+ (CRAC) channel. Together, these proteins participate in store-operated Ca2+ channel function. Defective regulation of intracellular Ca2+ is a hallmark of several diseases. In this review, we focus on Ca2+ regulation by the STIM1/Orai1 pathway and review evidence that implicates STIM1/Orai1 in several pathological conditions including cardiovascular and pulmonary diseases, among others.
Collapse
Affiliation(s)
- F R Giachini
- Department of Physiology, Georgia Heath Science University, Augusta, GA, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Liu W, Tang F, Chen J. Designing dynamical output feedback controllers for store-operated Ca²+ entry. Math Biosci 2010; 228:110-8. [PMID: 20816868 DOI: 10.1016/j.mbs.2010.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2009] [Revised: 06/21/2010] [Accepted: 08/27/2010] [Indexed: 12/26/2022]
Abstract
Store-operated calcium entry (SOCE) has been proposed as the main process controlling Ca²+ entry in non-excitable cells. Although recent breakthroughs in experimental studies of SOCE have been made, its mathematical modeling has not been developed. In the present work, SOCE is viewed as a feedback control system subject to an extracellular agonist disturbance and an extracellular calcium input. We then design a dynamic output feedback controller to reject the disturbance and track Ca²+ resting levels in the cytosol and the endoplasmic reticulum (ER). The constructed feedback control system is validated by published experimental data and its global asymptotic stability is proved by using the LaSalle's invariance principle. We then simulate the dynamic responses of STIM1 and Orai1, two major components in the operation of the store-operated channels, to the depletion of Ca²+ in the ER with thapsigargin, which show that: (1) Upon the depletion of Ca²+ in the ER, the concentrations of activated STIM1 and STIM1-Orai1 cluster are elevated gradually, indicating that STIM1 is accumulating in the ER-PM junctions and that the cytosolic portion of the active STIM1 is binding to Orai1 and driving the opening of CRAC channels for Ca²+ entry; (2) after the extracellular Ca²+ addition, the concentrations of both STIM1 and STIM1-Orai1 cluster decrease but still much higher than the original levels. We also simulate the system responses to the agonist disturbance, which show that, when a sequence of periodic agonist pulses is applied, the system returns to its equilibrium after each pulse. This indicates that the designed feedback controller can reject the disturbance and track the equilibrium.
Collapse
Affiliation(s)
- Weijiu Liu
- Department of Mathematics, University of Central Arkansas, 201 Donaghey Avenue, Conway, AR 72035, USA.
| | | | | |
Collapse
|
14
|
Shi Y, Song M, Guo R, Wang H, Gao P, Shi W, Huang L. Knockdown of stromal interaction molecule 1 attenuates hepatocyte growth factor-induced endothelial progenitor cell proliferation. Exp Biol Med (Maywood) 2010; 235:317-25. [PMID: 20404049 DOI: 10.1258/ebm.2009.009237] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Increased Ca(2+) entry through store-operated Ca(2+) channels (SOCCs) plays an essential role in the regulation of hepatocyte growth factor (HGF)-induced cell proliferation. Stromal interaction molecule 1 (STIM1) is thought to transmit endoplasmic reticulum (ER) Ca(2+) store depletion signals to the plasma membrane (PM), causing the opening of SOCCs in the PM. However, the relationship between HGF and STIM1 in endothelial progenitor cell (EPC) proliferation remains uncharacterized. The objective of this study was to evaluate the potential involvement of STIM1 in HGF-induced EPC proliferation. For this purpose, we used cultured rat bone marrow-derived EPCs and found that HGF-induced EPC proliferation at low concentrations. Store-operated Ca(2+) entry (SOCE) was elevated in HGF-treated EPCs, and the SOCC inhibitors 2-aminoethoxydiphenyl borate (2-APB) and BTP-2 inhibited the HGF-induced proliferation response. Moreover, STIM1 mRNA and protein expression levels were increased in response to HGF stimulation and knockdown of STMI1 decreased SOCE and prevented HGF-induced EPC proliferation. In conclusion, our data suggest that HGF-induced EPC proliferation is mediated partly via activation of STIM1.
Collapse
Affiliation(s)
- Yankun Shi
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|