1
|
Zhou S, Liu S, Jiang A, Li Z, Duan C, Li B. New insights into the stromal interaction molecule 2 function and its impact on the immunomodulation of tumor microenvironment. Cell Biosci 2024; 14:119. [PMID: 39272139 PMCID: PMC11395313 DOI: 10.1186/s13578-024-01292-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Immune cells-enhanced immunotherapy exhibits unprecedented overall survival-prolongation even curable in some cancer patients. Although so, most of the patients show no response. Tumor microenvironment (TME) where immune cells settle down has multi-faceted influences, but usually creates an immunosuppressive niche that facilitating tumor cells escape from immune attack. The metabolites and malnutrition of TME exert enormous effects on the resident immune cells, but the underlying mechanism is largely unknown. The stromal interaction molecules 2 (STIM2) is an endoplasmic reticulum (ER) calcium (Ca2+) sensor to maintain Ca2+ homeostasis. Notably, the cytosol STIM2 C-terminus is long with various domains that are available for the combination or/and molecular modification. This distinct structure endows STIM2 with a high susceptibility to numerous permeable physico-chemical molecules or protein interactions. STIM2 and its variants are extensively expressed in various immune cells, especially in T immune cells. STIM2 was reported closely correlated with the function of immune cells via regulating Ca2+ signaling, energy metabolism and cell fitness. Herein, we sum the latest findings on the STIM2 structure, focusing on its distinct characteristics and profound effect on the regulation of Ca2+ homeostasis and multi-talented functionality. We also outline the advancements on the underlying mechanism how STIM2 anomalies influence the function of immune cells and on the turbulent expression or/and amenably modification of STIM2 within the tumor niches. Then we discuss the translation of these researches into antitumor approaches, emphasizing the potential of STIM2 as a therapeutic target for direct inhibition of tumor cells or more activation towards immune cells driving to flare TME. This review is an update on STIM2, aiming to rationalize the potential of STIM2 as a therapeutic target for immunomodulation, engaging immune cells to exert the utmost anti-tumor effect.
Collapse
Affiliation(s)
- Shishan Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Shujie Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Anfeng Jiang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Zhiyuan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Bin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Serwach K, Nurowska E, Klukowska M, Zablocka B, Gruszczynska-Biegala J. STIM2 regulates NMDA receptor endocytosis that is induced by short-term NMDA receptor overactivation in cortical neurons. Cell Mol Life Sci 2023; 80:368. [PMID: 37989792 PMCID: PMC10663207 DOI: 10.1007/s00018-023-05028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/23/2023]
Abstract
Recent findings suggest an important role for the dysregulation of stromal interaction molecule (STIM) proteins, activators of store-operated Ca2+ channels, and the prolonged activation of N-methyl-D-aspartate receptors (NMDARs) in the development of neurodegenerative diseases. We previously demonstrated that STIM silencing increases Ca2+ influx through NMDAR and STIM-NMDAR2 complexes are present in neurons. However, the interplay between NMDAR subunits (GluN1, GluN2A, and GluN2B) and STIM1/STIM2 with regard to intracellular trafficking remains unknown. Here, we found that the activation of NMDAR endocytosis led to an increase in STIM2-GluN2A and STIM2-GluN2B interactions in primary cortical neurons. STIM1 appeared to migrate from synaptic to extrasynaptic sites. STIM2 silencing inhibited post-activation NMDAR translocation from the plasma membrane and synaptic spines and increased NMDAR currents. Our findings reveal a novel molecular mechanism by which STIM2 regulates NMDAR synaptic trafficking by promoting NMDAR endocytosis after receptor overactivation, which may suggest protection against excessive uncontrolled Ca2+ influx through NMDARs.
Collapse
Affiliation(s)
- Karolina Serwach
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Nurowska
- Department of Pharmacotherapy and Pharmaceutical Care, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Marta Klukowska
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Zablocka
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
3
|
Augustynek B, Gyimesi G, Dernič J, Sallinger M, Albano G, Klesse GJ, Kandasamy P, Grabmayr H, Frischauf I, Fuster DG, Peinelt C, Hediger MA, Bhardwaj R. Discovery of novel gating checkpoints in the Orai1 calcium channel by systematic analysis of constitutively active mutants of its paralogs and orthologs. Cell Calcium 2022; 105:102616. [PMID: 35792401 DOI: 10.1016/j.ceca.2022.102616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022]
Abstract
In humans, there are three paralogs of the Orai Ca2+ channel that form the core of the store-operated calcium entry (SOCE) machinery. While the STIM-mediated gating mechanism of Orai channels is still under active investigation, several artificial and natural variants are known to cause constitutive activity of the human Orai1 channel. Surprisingly, little is known about the conservation of the gating checkpoints among the different human Orai paralogs and orthologs in other species. In our work, we show that the mutation corresponding to the activating mutation H134A in transmembrane helix 2 (TM2) of human Orai1 also activates Orai2 and Orai3, likely via a similar mechanism. However, this cross-paralog conservation does not apply to the "ANSGA" nexus mutations in TM4 of human Orai1, which is reported to mimic the STIM1-activated state of the channel. In investigating the mechanistic background of these differences, we identified two positions, H171 and F246 in human Orai1, that are not conserved among paralogs and that seem to be crucial for the channel activation triggered by the "ANSGA" mutations in Orai1. However, mutations of the same residues still allow gating of Orai1 by STIM1, suggesting that the ANSGA mutant of Orai1 may not be a surrogate for the STIM1-activated state of the Orai1 channel. Our results shed new light on these important gating checkpoints and show that the gating mechanism of Orai channels is affected by multiple factors that are not necessarily conserved among orai homologs, such as the TM4-TM3 coupling.
Collapse
Affiliation(s)
- Bartłomiej Augustynek
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Gergely Gyimesi
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Jan Dernič
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Matthias Sallinger
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Giuseppe Albano
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Gabriel J Klesse
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Palanivel Kandasamy
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Herwig Grabmayr
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Irene Frischauf
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Daniel G Fuster
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Matthias A Hediger
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland.
| | - Rajesh Bhardwaj
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland; Current address: Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, 111 TW Alexander Drive, NC 27709, USA.
| |
Collapse
|
4
|
Collins HE, Zhang D, Chatham JC. STIM and Orai Mediated Regulation of Calcium Signaling in Age-Related Diseases. FRONTIERS IN AGING 2022; 3:876785. [PMID: 35821821 PMCID: PMC9261457 DOI: 10.3389/fragi.2022.876785] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/30/2022] [Indexed: 01/19/2023]
Abstract
Tight spatiotemporal regulation of intracellular Ca2+ plays a critical role in regulating diverse cellular functions including cell survival, metabolism, and transcription. As a result, eukaryotic cells have developed a wide variety of mechanisms for controlling Ca2+ influx and efflux across the plasma membrane as well as Ca2+ release and uptake from intracellular stores. The STIM and Orai protein families comprising of STIM1, STIM2, Orai1, Orai2, and Orai3, are evolutionarily highly conserved proteins that are core components of all mammalian Ca2+ signaling systems. STIM1 and Orai1 are considered key players in the regulation of Store Operated Calcium Entry (SOCE), where release of Ca2+ from intracellular stores such as the Endoplasmic/Sarcoplasmic reticulum (ER/SR) triggers Ca2+ influx across the plasma membrane. SOCE, which has been widely characterized in non-excitable cells, plays a central role in Ca2+-dependent transcriptional regulation. In addition to their role in Ca2+ signaling, STIM1 and Orai1 have been shown to contribute to the regulation of metabolism and mitochondrial function. STIM and Orai proteins are also subject to redox modifications, which influence their activities. Considering their ubiquitous expression, there has been increasing interest in the roles of STIM and Orai proteins in excitable cells such as neurons and myocytes. While controversy remains as to the importance of SOCE in excitable cells, STIM1 and Orai1 are essential for cellular homeostasis and their disruption is linked to various diseases associated with aging such as cardiovascular disease and neurodegeneration. The recent identification of splice variants for most STIM and Orai isoforms while complicating our understanding of their function, may also provide insight into some of the current contradictions on their roles. Therefore, the goal of this review is to describe our current understanding of the molecular regulation of STIM and Orai proteins and their roles in normal physiology and diseases of aging, with a particular focus on heart disease and neurodegeneration.
Collapse
Affiliation(s)
- Helen E. Collins
- Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Dingguo Zhang
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at Birmingham, Birmingham, AL, United States
| | - John C. Chatham
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: John C. Chatham,
| |
Collapse
|
5
|
Hunanyan L, Ghamaryan V, Makichyan A, Popugaeva E. Computer-Based Drug Design of Positive Modulators of Store-Operated Calcium Channels to Prevent Synaptic Dysfunction in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222413618. [PMID: 34948414 PMCID: PMC8707499 DOI: 10.3390/ijms222413618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 11/25/2022] Open
Abstract
Store-operated calcium entry (SOCE) constitutes a fine-tuning mechanism responsible for the replenishment of intracellular stores. Hippocampal SOCE is regulated by store-operated channels (SOC) organized in tripartite complex TRPC6/ORAI2/STIM2. It is suggested that in neurons, SOCE maintains intracellular homeostatic Ca2+ concentration at resting conditions and is needed to support the structure of dendritic spines. Recent evidence suggests that positive modulators of SOC are prospective drug candidates to treat Alzheimer’s disease (AD) at early stages. Although STIM2 and ORAI2 are definitely involved in the regulation of nSOC amplitude and a play major role in AD pathogenesis, growing evidence suggest that it is not easy to target these proteins pharmacologically. Existing positive modulators of TRPC6 are unsuitable for drug development due to either bad pharmacokinetics or side effects. Thus, we concentrate the review on perspectives to develop specific nSOC modulators based on available 3D structures of TRPC6, ORAI2, and STIM2. We shortly describe the structural features of existing models and the methods used to prepare them. We provide commonly used steps applied for drug design based on 3D structures of target proteins that might be used to develop novel AD preventing therapy.
Collapse
Affiliation(s)
- Lernik Hunanyan
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia;
- Laboratory of Structural Bioinformatics, Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia; (V.G.); (A.M.)
| | - Viktor Ghamaryan
- Laboratory of Structural Bioinformatics, Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia; (V.G.); (A.M.)
| | - Ani Makichyan
- Laboratory of Structural Bioinformatics, Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia; (V.G.); (A.M.)
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
6
|
Grabmayr H, Romanin C, Fahrner M. STIM Proteins: An Ever-Expanding Family. Int J Mol Sci 2020; 22:E378. [PMID: 33396497 PMCID: PMC7795233 DOI: 10.3390/ijms22010378] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/20/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
Stromal interaction molecules (STIM) are a distinct class of ubiquitously expressed single-pass transmembrane proteins in the endoplasmic reticulum (ER) membrane. Together with Orai ion channels in the plasma membrane (PM), they form the molecular basis of the calcium release-activated calcium (CRAC) channel. An intracellular signaling pathway known as store-operated calcium entry (SOCE) is critically dependent on the CRAC channel. The SOCE pathway is activated by the ligand-induced depletion of the ER calcium store. STIM proteins, acting as calcium sensors, subsequently sense this depletion and activate Orai ion channels via direct physical interaction to allow the influx of calcium ions for store refilling and downstream signaling processes. This review article is dedicated to the latest advances in the field of STIM proteins. New results of ongoing investigations based on the recently published functional data as well as structural data from nuclear magnetic resonance (NMR) spectroscopy and molecular dynamics (MD) simulations are reported and complemented with a discussion of the latest developments in the research of STIM protein isoforms and their differential functions in regulating SOCE.
Collapse
Affiliation(s)
| | - Christoph Romanin
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria;
| | - Marc Fahrner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria;
| |
Collapse
|
7
|
Novello MJ, Zhu J, Zhang M, Feng Q, Stathopulos PB. Synergistic stabilization by nitrosoglutathione-induced thiol modifications in the stromal interaction molecule-2 luminal domain suppresses basal and store operated calcium entry. Sci Rep 2020; 10:10177. [PMID: 32576932 PMCID: PMC7311479 DOI: 10.1038/s41598-020-66961-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/01/2020] [Indexed: 11/25/2022] Open
Abstract
Stromal interaction molecule−1 and −2 (STIM1/2) are endoplasmic reticulum (ER) membrane-inserted calcium (Ca2+) sensing proteins that, together with Orai1-composed Ca2+ channels on the plasma membrane (PM), regulate intracellular Ca2+ levels. Recent evidence suggests that S-nitrosylation of the luminal STIM1 Cys residues inhibits store operated Ca2+ entry (SOCE). However, the effects of thiol modifications on STIM2 during nitrosative stress and their role in regulating basal Ca2+ levels remain unknown. Here, we demonstrate that the nitric oxide (NO) donor nitrosoglutathione (GSNO) thermodynamically stabilizes the STIM2 Ca2+ sensing region in a Cys-specific manner. We uncovered a remarkable synergism in this stabilization involving the three luminal Cys of STIM2, which is unique to this paralog. S-Nitrosylation causes structural perturbations that converge on the face of the EF-hand and sterile α motif (EF-SAM) domain, implicated in unfolding-coupled activation. In HEK293T cells, enhanced free basal cytosolic Ca2+ and SOCE mediated by STIM2 overexpression could be attenuated by GSNO or mutation of the modifiable Cys located in the luminal domain. Collectively, we identify the Cys residues within the N-terminal region of STIM2 as modifiable targets during nitrosative stress that can profoundly and cooperatively affect basal Ca2+ and SOCE regulation.
Collapse
Affiliation(s)
- Matthew J Novello
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - Jinhui Zhu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London, Ontario, N6A5C1, Canada.,Dentistry, Schulich School of Medicine and Dentistry, the University of Western Ontario, London, Ontario, N6A5C1, Canada
| | - MengQi Zhang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London, Ontario, N6A5C1, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H8M5, Canada
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London, Ontario, N6A5C1, Canada.
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London, Ontario, N6A5C1, Canada.
| |
Collapse
|
8
|
Nelson HA, Roe MW. Molecular physiology and pathophysiology of stromal interaction molecules. Exp Biol Med (Maywood) 2018; 243:451-472. [PMID: 29363328 DOI: 10.1177/1535370218754524] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ca2+ release from the endoplasmic reticulum is an important component of Ca2+ signal transduction that controls numerous physiological processes in eukaryotic cells. Release of Ca2+ from the endoplasmic reticulum is coupled to the activation of store-operated Ca2+ entry into cells. Store-operated Ca2+ entry provides Ca2+ for replenishing depleted endoplasmic reticulum Ca2+ stores and a Ca2+ signal that regulates Ca2+-dependent intracellular biochemical events. Central to connecting discharge of endoplasmic reticulum Ca2+ stores following G protein-coupled receptor activation with the induction of store-operated Ca2+ entry are stromal interaction molecules (STIM1 and STIM2). These highly homologous endoplasmic reticulum transmembrane proteins function as sensors of the Ca2+ concentration within the endoplasmic reticulum lumen and activators of Ca2+ release-activated Ca2+ channels. Emerging evidence indicates that in addition to their role in Ca2+ release-activated Ca2+ channel gating and store-operated Ca2+ entry, STIM1 and STIM2 regulate other cellular signaling events. Recent studies have shown that disruption of STIM expression and function is associated with the pathogenesis of several diseases including autoimmune disorders, cancer, cardiovascular disease, and myopathies. Here, we provide an overview of the latest developments in the molecular physiology and pathophysiology of STIM1 and STIM2. Impact statement Intracellular Ca2+ signaling is a fundamentally important regulator of cell physiology. Recent studies have revealed that Ca2+-binding stromal interaction molecules (Stim1 and Stim2) expressed in the membrane of the endoplasmic reticulum (ER) are essential components of eukaryote Ca2+ signal transduction that control the activity of ion channels and other signaling effectors present in the plasma membrane. This review summarizes the most recent information on the molecular physiology and pathophysiology of stromal interaction molecules. We anticipate that the work presented in our review will provide new insights into molecular interactions that participate in interorganelle signaling crosstalk, cell function, and the pathogenesis of human diseases.
Collapse
Affiliation(s)
- Heather A Nelson
- 1 Department of Cell and Developmental Biology, 12302 SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael W Roe
- 1 Department of Cell and Developmental Biology, 12302 SUNY Upstate Medical University, Syracuse, NY 13210, USA.,2 Department of Medicine, 12302 SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
9
|
Song S, Carr SG, McDermott KM, Rodriguez M, Babicheva A, Balistrieri A, Ayon RJ, Wang J, Makino A, Yuan JXJ. STIM2 (Stromal Interaction Molecule 2)-Mediated Increase in Resting Cytosolic Free Ca 2+ Concentration Stimulates PASMC Proliferation in Pulmonary Arterial Hypertension. Hypertension 2018; 71:518-529. [PMID: 29358461 DOI: 10.1161/hypertensionaha.117.10503] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/11/2017] [Accepted: 12/27/2017] [Indexed: 12/13/2022]
Abstract
An increase in cytosolic free Ca2+ concentration ([Ca2+]cyt) in pulmonary artery smooth muscle cells (PASMCs) triggers pulmonary vasoconstriction and stimulates PASMC proliferation leading to vascular wall thickening. Here, we report that STIM2 (stromal interaction molecule 2), a Ca2+ sensor in the sarcoplasmic reticulum membrane, is required for raising the resting [Ca2+]cyt in PASMCs from patients with pulmonary arterial hypertension (PAH) and activating signaling cascades that stimulate PASMC proliferation and inhibit PASMC apoptosis. Downregulation of STIM2 in PAH-PASMCs reduces the resting [Ca2+]cyt, whereas overexpression of STIM2 in normal PASMCs increases the resting [Ca2+]cyt The increased resting [Ca2+]cyt in PAH-PASMCs is associated with enhanced phosphorylation (p) of CREB (cAMP response element-binding protein), STAT3 (signal transducer and activator of transcription 3), and AKT, increased NFAT (nuclear factor of activated T-cell) nuclear translocation, and elevated level of Ki67 (a marker of cell proliferation). Furthermore, the STIM2-associated increase in the resting [Ca2+]cyt also upregulates the antiapoptotic protein Bcl-2 in PAH-PASMCs. Downregulation of STIM2 in PAH-PASMCs with siRNA (1) decreases the level of pCREB, pSTAT3, and pAKT and inhibits NFAT nuclear translocation, thereby attenuating proliferation, and (2) decreases Bcl-2, which leads to an increase of apoptosis. In summary, these data indicate that upregulated STIM2 in PAH-PASMCs, by raising the resting [Ca2+]cyt, contributes to enhancing PASMC proliferation by activating the CREB, STAT3, AKT, and NFAT signaling pathways and stimulating PASMC proliferation. The STIM2-associated increase in the resting [Ca2+]cyt is also involved in upregulating Bcl-2 that makes PAH-PASMCs resistant to apoptosis, and thus plays an important role in sustained pulmonary vasoconstriction and excessive pulmonary vascular remodeling in patients with PAH.
Collapse
Affiliation(s)
- Shanshan Song
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Shane G Carr
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Kimberly M McDermott
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Marisela Rodriguez
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Aleksandra Babicheva
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Angela Balistrieri
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Ramon J Ayon
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Jian Wang
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Ayako Makino
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson
| | - Jason X-J Yuan
- From the Division of Translational and Regenerative Medicine, Department of Medicine (S.S., S.G.C., K.M.M., M.R., A. Babicheva, A. Balistrieri, R.J.A., J.W., A.M., J.X.-J.Y.) and Department of Physiology (A.M., J.X.-J.Y.), The University of Arizona College of Medicine, Tucson.
| |
Collapse
|
10
|
Zhu J, Feng Q, Stathopulos PB. The STIM-Orai Pathway: STIM-Orai Structures: Isolated and in Complex. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:15-38. [PMID: 28900907 DOI: 10.1007/978-3-319-57732-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Considerable progress has been made elucidating the molecular mechanisms of calcium (Ca2+) sensing by stromal interaction molecules (STIMs) and the basis for Orai channel activity. This chapter focuses on the available high-resolution structural details of STIM and Orai proteins with respect to the regulation of store-operated Ca2+ entry (SOCE). Solution structures of the Ca2+-sensing domains of STIM1 and STIM2 are reviewed in detail, crystal structures of cytosolic coiled-coil STIM fragments are discussed, and an overview of the closed Drosophila melanogaster Orai hexameric structure is provided. Additionally, we highlight structures of human Orai1 N-terminal and C-terminal domains in complex with calmodulin and human STIM1, respectively. Ultimately, the accessible structural data are discussed in terms of potential mechanisms of action and cohesiveness with functional observations.
Collapse
Affiliation(s)
- Jinhui Zhu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5C1
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5C1
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5C1.
| |
Collapse
|
11
|
Mignen O, Constantin B, Potier-Cartereau M, Penna A, Gautier M, Guéguinou M, Renaudineau Y, Shoji KF, Félix R, Bayet E, Buscaglia P, Debant M, Chantôme A, Vandier C. Constitutive calcium entry and cancer: updated views and insights. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:395-413. [PMID: 28516266 DOI: 10.1007/s00249-017-1216-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 03/10/2017] [Accepted: 04/26/2017] [Indexed: 12/20/2022]
Abstract
Tight control of basal cytosolic Ca2+ concentration is essential for cell survival and to fine-tune Ca2+-dependent cell functions. A way to control this basal cytosolic Ca2+ concentration is to regulate membrane Ca2+ channels including store-operated Ca2+ channels and secondary messenger-operated channels linked to G-protein-coupled or tyrosine kinase receptor activation. Orai, with or without its reticular STIM partner and Transient Receptor Potential (TRP) proteins, were considered to be the main Ca2+ channels involved. It is well accepted that, in response to cell stimulation, opening of these Ca2+ channels contributes to Ca2+ entry and the transient increase in cytosolic Ca2+ concentration involved in intracellular signaling. However, in various experimental conditions, Ca2+ entry and/or Ca2+ currents can be recorded at rest, without application of any experimental stimulation. This led to the proposition that some plasma membrane Ca2+ channels are already open/activated in basal condition, contributing therefore to constitutive Ca2+ entry. This article focuses on direct and indirect observations supporting constitutive activity of channels belonging to the Orai and TRP families and on the mechanisms underlying their basal/constitutive activities.
Collapse
Affiliation(s)
- Olivier Mignen
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Bruno Constantin
- STIM, ERL 7368 CNRS Université de Poitiers, Poitiers, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Marie Potier-Cartereau
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Aubin Penna
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Mathieu Gautier
- EA4667, Université de Picardie Jules Verne, 80039, Amiens, France
| | - Maxime Guéguinou
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Yves Renaudineau
- EA 2216, Inserm ESPRI, ERI 29, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Kenji F Shoji
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Romain Félix
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Elsa Bayet
- STIM, ERL 7368 CNRS Université de Poitiers, Poitiers, France
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Paul Buscaglia
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Marjolaine Debant
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- EA 2216, Inserm ESPRI, ERI 29, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Aurélie Chantôme
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Christophe Vandier
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France.
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France.
| |
Collapse
|
12
|
Berna-Erro A, Jardin I, Salido GM, Rosado JA. Role of STIM2 in cell function and physiopathology. J Physiol 2017; 595:3111-3128. [PMID: 28087881 DOI: 10.1113/jp273889] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/03/2017] [Indexed: 01/01/2023] Open
Abstract
An endoplasmic reticulum (ER)-resident protein that regulates cytosolic and ER free-Ca2+ concentration by induction of store-operated calcium entry: that is the original definition of STIM2 and its function. While its activity strongly depends on the amount of calcium stored in the ER, its function goes further, to intracellular signalling and gene expression. Initially under-studied owing to the prominent function of STIM1, STIM2 came to be regarded as vital in mice, gradually emerging as an important player in the nervous system, and cooperating with STIM1 in the immune system. STIM2 has also been proposed as a relevant player in pathological conditions related to ageing, Alzheimer's and Huntington's diseases, autoimmune disorders and cancer. The discovery of additional functions, together with new splicing forms with opposite roles, has clarified existing controversies about STIM2 function in SOCE. With STIM2 being essential for life, but apparently not for development, newly available data demonstrate a complex and still intriguing behaviour that this review summarizes, updating current knowledge of STIM2 function.
Collapse
Affiliation(s)
- Alejandro Berna-Erro
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Isaac Jardin
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003, Cáceres, Spain
| | - Gines M Salido
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003, Cáceres, Spain
| |
Collapse
|
13
|
Niemeyer BA. The STIM-Orai Pathway: Regulation of STIM and Orai by Thiol Modifications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:99-116. [PMID: 28900911 DOI: 10.1007/978-3-319-57732-6_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cysteines are among the least abundant amino acids found in proteins. Due to their unique nucleophilic thiol group, they are able to undergo a broad range of chemical modifications besides their known role in disulfide formation, such as S-sulfenylation (-SOH), S-sulfinylation (-SO(2)H), S-sufonylation (-SO(3)H), S-glutathionylation (-SSG), and S-sulfhydration (-SSH), among others. These posttranslational modifications can be irreversible and act as transitional modifiers or as reversible on-off switches for the function of proteins. Disturbances of the redox homeostasis, for example, in situations of increased oxidative stress, can contribute to a range of diseases. Because Ca2+ signaling mediated by store-operated calcium entry (SOCE) is involved in a plethora of cellular responses, the cross-talk between reactive oxygen species (ROS) and Ca2+ is critical for homeostatic control. Identification of calcium regulatory protein targets of thiol redox modifications is needed to understand their role in biology and disease.
Collapse
Affiliation(s)
- Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany.
| |
Collapse
|
14
|
Bhardwaj R, Hediger MA, Demaurex N. Redox modulation of STIM-ORAI signaling. Cell Calcium 2016; 60:142-52. [DOI: 10.1016/j.ceca.2016.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 12/14/2022]
|
15
|
Gao X, Xia J, Munoz FM, Manners MT, Pan R, Meucci O, Dai Y, Hu H. STIMs and Orai1 regulate cytokine production in spinal astrocytes. J Neuroinflammation 2016; 13:126. [PMID: 27245842 PMCID: PMC4886427 DOI: 10.1186/s12974-016-0594-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/23/2016] [Indexed: 12/30/2022] Open
Abstract
Background Our previous study demonstrated that a store-operated calcium channel (SOCC) inhibitor (YM-58483) has central analgesic effects. However, the cellular and molecular mechanisms of such effects remain to be determined. It is well-known that glial cells play important roles in central sensitization. SOC entry (SOCE) has been implicated in many cell types including cortical astrocytes. However, the role of the SOCC family in the function of astrocytes has not been determined. Here, we thoroughly investigated the expression and the functional significance of SOCCs in spinal astrocytes. Methods Primary cultured astrocytes were prepared from neonatal (P2–P3) CD1 mice. Expressions of mRNAs and proteins were respectively assessed by real-time PCR and Western blot analysis. SOCE was measured using a calcium imaging system. Live-cell STIM1 translocation was detected using a confocal microscope. Cytokine levels were measured by the enzyme-linked immunosorbent assay. Results We found that the SOCC family is expressed in spinal astrocytes and that depletion of calcium stores from the endoplasmic reticulum by cyclopiazonic acid (CPA) resulted in a large sustained calcium entry, which was blocked by SOCC inhibitors. Using the siRNA knockdown approach, we identified STIM1 and Orai1 as primary components of SOCCs in spinal astrocytes. We also observed thapsigargin (TG)- or CPA-induced puncta formation of STIM1 and Orai1. In addition, activation of SOCCs remarkably promoted TNF-α and IL-6 production in spinal astrocytes, which were greatly attenuated by knockdown of STIM1 or Orai1. Importantly, knockdown of STIM2 and Orai1 dramatically decreased lipopolysaccharide-induced TNF-α and IL-6 production without changing cell viability. Conclusions This study presents the first evidence that STIM1, STIM2, and Orai1 mediate SOCE and are involved in cytokine production in spinal astrocytes. Our findings provide the basis for future assessment of SOCCs in pain and other central nervous system disorders associated with abnormal astrocyte activities.
Collapse
Affiliation(s)
- Xinghua Gao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.,Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Jingsheng Xia
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Frances M Munoz
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Melissa T Manners
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Rong Pan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Huijuan Hu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
16
|
Niemeyer BA. Changing calcium: CRAC channel (STIM and Orai) expression, splicing, and posttranslational modifiers. Am J Physiol Cell Physiol 2016; 310:C701-9. [PMID: 26911279 DOI: 10.1152/ajpcell.00034.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A wide variety of cellular function depends on the dynamics of intracellular Ca(2+) signals. Especially for relatively slow and lasting processes such as gene expression, cell proliferation, and often migration, cells rely on the store-operated Ca(2+) entry (SOCE) pathway, which is particularly prominent in immune cells. SOCE is initiated by the sensor proteins (STIM1, STIM2) located within the endoplasmic reticulum (ER) registering the Ca(2+) concentration within the ER, and upon its depletion, cluster and trap Orai (Orai1-3) proteins located in the plasma membrane (PM) into ER-PM junctions. These regions become sites of highly selective Ca(2+) entry predominantly through Orai1-assembled channels, which, among other effector functions, is necessary for triggering NFAT translocation into the nucleus. What is less clear is how the spatial and temporal spread of intracellular Ca(2+) is shaped and regulated by differential expression of the individual SOCE genes and their splice variants, their heteromeric combinations and pre- and posttranslational modifications. This review focuses on principle mechanisms regulating expression, splicing, and targeting of Ca(2+) release-activated Ca(2+) (CRAC) channels.
Collapse
Affiliation(s)
- Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
17
|
Majewski L, Kuznicki J. SOCE in neurons: Signaling or just refilling? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1940-52. [DOI: 10.1016/j.bbamcr.2015.01.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 01/14/2023]
|
18
|
Moccia F, Zuccolo E, Soda T, Tanzi F, Guerra G, Mapelli L, Lodola F, D'Angelo E. Stim and Orai proteins in neuronal Ca(2+) signaling and excitability. Front Cell Neurosci 2015; 9:153. [PMID: 25964739 PMCID: PMC4408853 DOI: 10.3389/fncel.2015.00153] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/03/2015] [Indexed: 02/01/2023] Open
Abstract
Stim1 and Orai1 are ubiquitous proteins that have long been known to mediate Ca2+ release-activated Ca2+ (CRAC) current (ICRAC) and store-operated Ca2+ entry (SOCE) only in non-excitable cells. SOCE is activated following the depletion of the endogenous Ca2+ stores, which are mainly located within the endoplasmic reticulum (ER), to replete the intracellular Ca2+ reservoir and engage specific Ca2+-dependent processes, such as proliferation, migration, cytoskeletal remodeling, and gene expression. Their paralogs, Stim2, Orai2 and Orai3, support SOCE in heterologous expression systems, but their physiological role is still obscure. Ca2+ inflow in neurons has long been exclusively ascribed to voltage-operated and receptor-operated channels. Nevertheless, recent work has unveiled that Stim1–2 and Orai1-2, but not Orai3, proteins are also expressed and mediate SOCE in neurons. Herein, we survey current knowledge about the neuronal distribution of Stim and Orai proteins in rodent and human brains; we further discuss that Orai2 is the main pore-forming subunit of CRAC channels in central neurons, in which it may be activated by either Stim1 or Stim2 depending on species, brain region and physiological stimuli. We examine the functions regulated by SOCE in neurons, where this pathway is activated under resting conditions to refill the ER, control spinogenesis and regulate gene transcription. Besides, we highlighted the possibility that SOCE also controls neuronal excitation and regulate synaptic plasticity. Finally, we evaluate the involvement of Stim and Orai proteins in severe neurodegenerative and neurological disorders, such as Alzheimer’s disease and epilepsy.
Collapse
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Teresa Soda
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy
| | - Franco Tanzi
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Lisa Mapelli
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Roma, Italy
| | - Francesco Lodola
- Laboratory of Molecular Cardiology, IRCCS Fondazione Salvatore Maugeri Pavia, Italy
| | - Egidio D'Angelo
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Brain Connectivity Center, C. Mondino National Neurological Institute, Fondazione IRCCS Policlinico San Matteo Pavia Pavia, Italy
| |
Collapse
|
19
|
Miederer AM, Alansary D, Schwär G, Lee PH, Jung M, Helms V, Niemeyer BA. A STIM2 splice variant negatively regulates store-operated calcium entry. Nat Commun 2015; 6:6899. [PMID: 25896806 PMCID: PMC4411291 DOI: 10.1038/ncomms7899] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/11/2015] [Indexed: 02/01/2023] Open
Abstract
Cellular homeostasis relies upon precise regulation of Ca(2+) concentration. Stromal interaction molecule (STIM) proteins regulate store-operated calcium entry (SOCE) by sensing Ca(2+) concentration in the ER and forming oligomers to trigger Ca(2+) entry through plasma membrane-localized Orai1 channels. Here we characterize a STIM2 splice variant, STIM2.1, which retains an additional exon within the region encoding the channel-activating domain. Expression of STIM2.1 is ubiquitous but its abundance relative to the more common STIM2.2 variant is dependent upon cell type and highest in naive T cells. STIM2.1 knockdown increases SOCE in naive CD4(+) T cells, whereas knockdown of STIM2.2 decreases SOCE. Conversely, overexpression of STIM2.1, but not STIM2.2, decreases SOCE, indicating its inhibitory role. STIM2.1 interaction with Orai1 is impaired and prevents Orai1 activation, but STIM2.1 shows increased affinity towards calmodulin. Our results imply STIM2.1 as an additional player tuning Orai1 activation in vivo.
Collapse
Affiliation(s)
- Anna-Maria Miederer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, Homburg 66421, Germany
| | - Dalia Alansary
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, Homburg 66421, Germany
| | - Gertrud Schwär
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, Homburg 66421, Germany
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Po-Hsien Lee
- Center for Bioinformatics, Saarland University, Campus E2 1, R. 315, PO Box 151150, Saarbrücken 66041, Germany
| | - Martin Jung
- Department of Medical Biochemistry and Molecular Biology, Saarland University, Building 44, Homburg 66421, Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Campus E2 1, R. 315, PO Box 151150, Saarbrücken 66041, Germany
| | - Barbara A. Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, Homburg 66421, Germany
| |
Collapse
|
20
|
Hoth M, Niemeyer BA. The neglected CRAC proteins: Orai2, Orai3, and STIM2. CURRENT TOPICS IN MEMBRANES 2014; 71:237-71. [PMID: 23890118 DOI: 10.1016/b978-0-12-407870-3.00010-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Plasma-membrane-localized Orai1 ion channel subunits interacting with ER-localized STIM1 molecules comprise the major subunit composition responsible for calcium release-activated calcium channels. STIM1 "translates" the Ca(2+) store content into Orai1 activity, making it a store-operated channel. Surprisingly, in addition to being the physical activator, STIM1 also modulates Orai1 properties, including its inactivation and permeation (see Chapter 1). STIM1 is thus more than a pure Orai1 activator. Within the past 7 years following the discovery of STIM and Orai proteins, the molecular mechanisms of STIM1/Orai1 activity and their functional importance have been studied in great detail. Much less is currently known about the other isoforms STIM2, Orai2, and Orai3. In this chapter, we summarize the current knowledge about STIM2, Orai2, and Orai3 properties and function. Are these homologues mainly modulators of predominantly STIM1/Orai1-mediated complexes or do store-dependent or -independent functions such as regulation of basal Ca(2+) concentration and activation of Orai3-containing complexes by arachidonic acid or by estrogen receptors point toward their "true" physiological function? Is Orai2 the Orai1 of neurons? A major focus of the review is on the functional relevance of STIM2, Orai2, and Orai3, some of which still remains speculative.
Collapse
Affiliation(s)
- Markus Hoth
- Department of Biophysics, Saarland University, Homburg, Germany
| | | |
Collapse
|
21
|
Gruszczynska-Biegala J, Kuznicki J. Native STIM2 and ORAI1 proteins form a calcium-sensitive and thapsigargin-insensitive complex in cortical neurons. J Neurochem 2013; 126:727-38. [PMID: 23711249 DOI: 10.1111/jnc.12320] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/30/2013] [Accepted: 05/17/2013] [Indexed: 01/14/2023]
Abstract
In non-excitatory cells, stromal interaction molecule 1 (STIM1) and STIM2 mediate store-operated calcium entry via an interaction with ORAI1 calcium channels. However, in neurons, STIM2 over-expression appears to play a role in calcium homeostasis that is different from STIM1 over-expression. The aim of this study was to establish the role and localization of native STIM2 in the neuronal cell. Co-immunoprecipitation experiments revealed that the interaction between endogenous STIM2 and ORAI1 was greater in a low-calcium medium than in a high-calcium medium. Using a Proximity Ligation Assay (PLA), the number of apparent complexes of endogenous STIM2 with ORAI1 was quantified. No change in the number of PLA signals was observed in the presence of thapsigargin, which depletes calcium from the endoplasmic reticulum (ER). However, the number of apparent STIM2-ORAI1 complexes increased when intracellular and subsequently ER calcium concentrations were decreased by BAPTA-AM or a low-calcium medium. Both Fura-2 acetoxymethyl ester calcium imaging and PLA in the same neuronal cell indicated that the calcium responses correlated strongly with the number of endogenous STIM2-ORAI1 complexes. The small drop in calcium levels in the ER caused by decreased intracellular calcium levels appeared to initiate the calcium-sensitive and thapsigargin-insensitive interaction between STIM2 and ORAI1. We show in neuronal somata the formation of endogenous complexes of stromal interaction molecule 2 (STIM2) with ORAI1 calcium channels. Their number increased when intracellular Ca²⁺ concentrations were decreased by the Ca²⁺ chelator BAPTA-AM or a low-calcium medium (EGTA), but did not in the presence of thapsigargin (TG). We conclude that the small drop of Ca²⁺ level in endoplasmic reticulum, due to the decreased level of intracellular Ca²⁺, is sufficient to trigger STIM2-ORAI1 complex formation in a thapsigargin-insensitive manner.
Collapse
|
22
|
O'Leary EE, Mazurkiewicz-Muñoz AM, Argetsinger LS, Maures TJ, Huynh HT, Carter-Su C. Identification of steroid-sensitive gene-1/Ccdc80 as a JAK2-binding protein. Mol Endocrinol 2013; 27:619-34. [PMID: 23449887 DOI: 10.1210/me.2011-1275] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The tyrosine kinase Janus kinase 2 (JAK2) is activated by many cytokine receptors, including receptors for GH, leptin, and erythropoietin. However, very few proteins have been identified as binding partners for JAK2. Using a yeast 2-hybrid screen, we identified steroid-sensitive gene-1 (SSG1)/coiled-coil domain-containing protein 80 (Ccdc80) as a JAK2-binding partner. We demonstrate that Ccdc80 preferentially binds activated, tyrosyl-phosphorylated JAK2 but not kinase-inactive JAK2 (K882E) in both yeast and mammalian systems. Ccdc80 is tyrosyl phosphorylated in the presence of JAK2. The binding of Ccdc80 to JAK2 occurs via 1 or more of the 3 DUDES/SRPX (DRO1-URB-DRS-Equarin-SRPUL/sushi repeat containing protein, x-linked) domain 5 domains of Ccdc80. Mutagenesis of the second DUDES domain suggests that the N-terminal third of the DUDES domain is sufficient for JAK2 binding. Ccdc80 does not alter the kinase activity of JAK2. However, Ccdc80 increases GH-dependent phosphorylation of Stat (signal transducer and activator of transcription) 5b on Tyr699 and substantially enhances both basal and GH-dependent phosphorylation/activation of Stat3 on Tyr705. Furthermore, Ccdc80 belongs to the group of proteins that function both in the intracellular compartment and are secreted. Secreted Ccdc80 associates with the extracellular matrix and is also found in the medium. A substantial portion of the Ccdc80 detected in the medium is cleaved. Finally, consistent with the DUDES domain serving as a JAK2-binding domain, we also demonstrate that another protein that contains a DUDES domain, SRPX2, binds preferentially to the activated tyrosyl-phosphorylated form of JAK2.
Collapse
Affiliation(s)
- Erin E O'Leary
- Graduate Program in Cellular and Molecular Biology, The University of Michigan Medical School, Ann Arbor, Michigan 48109-5622, USA
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Ca2+i signalling is pivotal to sperm function. Progesterone, the best-characterized agonist of human sperm Ca2+i signalling, stimulates a biphasic [Ca2+]i rise, comprising a transient and subsequent sustained phase. In accordance with recent reports that progesterone directly activates CatSper, the [Ca2+]i transient was detectable in the anterior flagellum (where CatSper is expressed) 1–2 s before responses in the head and neck. Pre-treatment with 5 μM 2-APB (2-aminoethoxydiphenyl borate), which enhances activity of store-operated channel proteins (Orai) by facilitating interaction with their activator [STIM (stromal interaction molecule)] ‘amplified’ progesterone-induced [Ca2+]i transients at the sperm neck/midpiece without modifying kinetics. The flagellar [Ca2+]i response was unchanged. 2-APB (5 μM) also enhanced the sustained response in the midpiece, possibly reflecting mitochondrial Ca2+ accumulation downstream of the potentiated [Ca2+]i transient. Pre-treatment with 50–100 μM 2-APB failed to potentiate the transient and suppressed sustained [Ca2+]i elevation. When applied during the [Ca2+]i plateau, 50–100 μM 2-APB caused a transient fall in [Ca2+]i, which then recovered despite the continued presence of 2-APB. Loperamide (a chemically different store-operated channel agonist) enhanced the progesterone-induced [Ca2+]i signal and potentiated progesterone-induced hyperactivated motility. Neither 2-APB nor loperamide raised pHi (which would activate CatSper) and both compounds inhibited CatSper currents. STIM and Orai were detected and localized primarily to the neck/midpiece and acrosome where Ca2+ stores are present and the effects of 2-APB are focussed, but store-operated currents could not be detected in human sperm. We propose that 2-APB-sensitive channels amplify [Ca2+]i elevation induced by progesterone (and other CatSper agonists), amplifying, propagating and providing spatio-temporal complexity in [Ca2+]i signals of human sperm.
Collapse
|
24
|
Abstract
Store-operated calcium (Ca(2+)) entry (SOCE) is a vital Ca(2+) signaling pathway in nonexcitable as well as electrically excitable cells, regulating countless physiological and pathophysiological pathways. Stromal interaction molecules (STIMs) are the principal regulating molecules of SOCE, sensing changes in sarco-/endoplasmic reticulum (S/ER) luminal Ca(2+) levels and directly interacting with the Orai channel subunits to orchestrate the opening of Ca(2+) release-activated Ca(2+) (CRAC) channels. Recent atomic resolution structures on human STIM1 and STIM2 have illuminated critical mechanisms of STIM function in SOCE; further, the first high-resolution structure of the Drosophila melanogaster Orai channel has revealed vital data on the atomic composition of the CRAC channel pore and the assembly of individual Orai subunits. This chapter focuses on the mechanistic information garnered from these high-resolution structures and the supporting biophysical, biochemical, and live cell work that has enhanced our understanding of the relationship between STIM and Orai structural features and CRAC channel function.
Collapse
|
25
|
Davis FM, Peters AA, Grice DM, Cabot PJ, Parat MO, Roberts-Thomson SJ, Monteith GR. Non-stimulated, agonist-stimulated and store-operated Ca2+ influx in MDA-MB-468 breast cancer cells and the effect of EGF-induced EMT on calcium entry. PLoS One 2012; 7:e36923. [PMID: 22666335 PMCID: PMC3364242 DOI: 10.1371/journal.pone.0036923] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 04/17/2012] [Indexed: 12/30/2022] Open
Abstract
In addition to their well-defined roles in replenishing depleted endoplasmic reticulum (ER) Ca2+ reserves, molecular components of the store-operated Ca2+ entry pathway regulate breast cancer metastasis. A process implicated in cancer metastasis that describes the conversion to a more invasive phenotype is epithelial-mesenchymal transition (EMT). In this study we show that EGF-induced EMT in MDA-MB-468 breast cancer cells is associated with a reduction in agonist-stimulated and store-operated Ca2+ influx, and that MDA-MB-468 cells prior to EMT induction have a high level of non-stimulated Ca2+ influx. The potential roles for specific Ca2+ channels in these pathways were assessed by siRNA-mediated silencing of ORAI1 and transient receptor potential canonical type 1 (TRPC1) channels in MDA-MB-468 breast cancer cells. Non-stimulated, agonist-stimulated and store-operated Ca2+ influx were significantly inhibited with ORAI1 silencing. TRPC1 knockdown attenuated non-stimulated Ca2+ influx in a manner dependent on Ca2+ influx via ORAI1. TRPC1 silencing was also associated with reduced ERK1/2 phosphorylation and changes in the rate of Ca2+ release from the ER associated with the inhibition of the sarco/endoplasmic reticulum Ca2+-ATPase (time to peak [Ca2+]CYT = 188.7±34.6 s (TRPC1 siRNA) versus 124.0±9.5 s (non-targeting siRNA); P<0.05). These studies indicate that EMT in MDA-MB-468 breast cancer cells is associated with a pronounced remodeling of Ca2+ influx, which may be due to altered ORAI1 and/or TRPC1 channel function. Our findings also suggest that TRPC1 channels in MDA-MB-468 cells contribute to ORAI1-mediated Ca2+ influx in non-stimulated cells.
Collapse
Affiliation(s)
- Felicity M. Davis
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Amelia A. Peters
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Desma M. Grice
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Peter J. Cabot
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Marie-Odile Parat
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Gregory R. Monteith
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
26
|
Bogeski I, Kilch T, Niemeyer BA. ROS and SOCE: recent advances and controversies in the regulation of STIM and Orai. J Physiol 2012; 590:4193-200. [PMID: 22615429 DOI: 10.1113/jphysiol.2012.230565] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Store-operated Ca(2+) entry (SOCE) is a widespread mechanism in cells to raise cytosolic Ca(2+) and to refill Ca(2+) stores. T cells critically rely on SOCE mediated by stromal interaction molecules (STIM) and Orai molecules for their activation and regulation of gene transcription; cells such as muscle cells, neurons or melanocytes probably utilize SOCE for the transmission of inducible receptor-mediated function as well as for generalized Ca(2+) homeostasis mechanisms. Exposure to environmental or cell-intrinisic reactive oxygen species (ROS) can affect several components involved in Ca(2+) homeostasis and thus alter multiple pathways. While all cells have a capacity to produce intracellular ROS, exposure of immune and skin cells to extracellular oxidative stress is particularly high during inflammation and/or with UV exposure. This review briefly summarizes cell-intrinsic sources of ROS and focuses on current findings and controversies regarding the regulation of STIM and Orai by oxidative modifications. We also introduce melanocytes as a new model system to study the function of STIM and Orai isoforms under physiological conditions that include exposure to UV light as an activating stimulus.
Collapse
Affiliation(s)
- Ivan Bogeski
- Department of Biophysics, Saarland University, 66421 Homburg, Germany
| | | | | |
Collapse
|
27
|
López E, Salido GM, Rosado JA, Berna-Erro A. Unraveling STIM2 function. J Physiol Biochem 2012; 68:619-33. [PMID: 22477146 DOI: 10.1007/s13105-012-0163-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 03/13/2012] [Indexed: 12/11/2022]
Abstract
The discovery of molecular players in capacitative calcium (Ca(2+)) entry, also referred to as store-operated Ca(2+) entry (SOCE), supposed a great advance in the knowledge of cellular mechanisms of Ca(2+) entry, which are essential for a broad range of cellular functions. The identification of STIM1 and STIM2 proteins as the sensors of Ca(2+) stored in the endoplasmic reticulum unraveled the mechanism by which depletion of intracellular Ca(2+) stores is communicated to store-operated Ca(2+) channels located in the plasma membrane, triggering the activation of SOCE and intracellular Ca(2+)-dependent signaling cascades. Initial studies suggested a dominant function of STIM1 in SOCE and SOCE-dependent cellular functions compared to STIM2, especially those that participate in immune responses. Consequently, most of the subsequent studies focused on STIM1. However, during the last years, STIM2 has been demonstrated to play a more relevant and complex function than initially reported, being even important to sustain normal life in mice. These studies have led to reconsider the role of STIM2 in SOCE and its relevance in cellular physiology. This review is intended to summarize and provide an overview of the current data available about this exciting isoform, STIM2, and its actual position together with STIM1 in the mechanism of SOCE.
Collapse
Affiliation(s)
- Esther López
- Department of Physiology (Cellular Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | | | | | | |
Collapse
|
28
|
Lee JM, Davis FM, Roberts-Thomson SJ, Monteith GR. Ion channels and transporters in cancer. 4. Remodeling of Ca(2+) signaling in tumorigenesis: role of Ca(2+) transport. Am J Physiol Cell Physiol 2011; 301:C969-76. [PMID: 21593447 DOI: 10.1152/ajpcell.00136.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Ca(2+) signal has major roles in cellular processes important in tumorigenesis, including migration, invasion, proliferation, and apoptotic sensitivity. New evidence has revealed that, aside from altered expression and effects on global cytosolic free Ca(2+) levels via direct transport of Ca(2+), some Ca(2+) pumps and channels are able to contribute to tumorigenesis via mechanisms that are independent of their ability to transport Ca(2+) or effect global Ca(2+) homeostasis in the cytoplasm. Here, we review some of the most recent studies that present evidence of altered Ca(2+) channel or pump expression in tumorigenesis and discuss the importance and complexity of localized Ca(2+) signaling in events critical for tumor formation.
Collapse
Affiliation(s)
- Jane M Lee
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|