1
|
Liang X, Zhang N, Pan H, Xie J, Han W. Development of Store-Operated Calcium Entry-Targeted Compounds in Cancer. Front Pharmacol 2021; 12:688244. [PMID: 34122115 PMCID: PMC8194303 DOI: 10.3389/fphar.2021.688244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is the major pathway of Ca2+ entry in mammalian cells, and regulates a variety of cellular functions including proliferation, motility, apoptosis, and death. Accumulating evidence has indicated that augmented SOCE is related to the generation and development of cancer, including tumor formation, proliferation, angiogenesis, metastasis, and antitumor immunity. Therefore, the development of compounds targeting SOCE has been proposed as a potential and effective strategy for use in cancer therapy. In this review, we summarize the current research on SOCE inhibitors and blockers, discuss their effects and possible mechanisms of action in cancer therapy, and induce a new perspective on the treatment of cancer.
Collapse
Affiliation(s)
- Xiaojing Liang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ningxia Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Villalobo A, Berchtold MW. The Role of Calmodulin in Tumor Cell Migration, Invasiveness, and Metastasis. Int J Mol Sci 2020; 21:ijms21030765. [PMID: 31991573 PMCID: PMC7037201 DOI: 10.3390/ijms21030765] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Calmodulin (CaM) is the principal Ca2+ sensor protein in all eukaryotic cells, that upon binding to target proteins transduces signals encoded by global or subcellular-specific changes of Ca2+ concentration within the cell. The Ca2+/CaM complex as well as Ca2+-free CaM modulate the activity of a vast number of enzymes, channels, signaling, adaptor and structural proteins, and hence the functionality of implicated signaling pathways, which control multiple cellular functions. A basic and important cellular function controlled by CaM in various ways is cell motility. Here we discuss the role of CaM-dependent systems involved in cell migration, tumor cell invasiveness, and metastasis development. Emphasis is given to phosphorylation/dephosphorylation events catalyzed by myosin light-chain kinase, CaM-dependent kinase-II, as well as other CaM-dependent kinases, and the CaM-dependent phosphatase calcineurin. In addition, the role of the CaM-regulated small GTPases Rac1 and Cdc42 (cell division cycle protein 42) as well as CaM-binding adaptor/scaffold proteins such as Grb7 (growth factor receptor bound protein 7), IQGAP (IQ motif containing GTPase activating protein) and AKAP12 (A kinase anchoring protein 12) will be reviewed. CaM-regulated mechanisms in cancer cells responsible for their greater migratory capacity compared to non-malignant cells, invasion of adjacent normal tissues and their systemic dissemination will be discussed, including closely linked processes such as the epithelial–mesenchymal transition and the activation of metalloproteases. This review covers as well the role of CaM in establishing metastatic foci in distant organs. Finally, the use of CaM antagonists and other blocking techniques to downregulate CaM-dependent systems aimed at preventing cancer cell invasiveness and metastasis development will be outlined.
Collapse
Affiliation(s)
- Antonio Villalobo
- Cancer and Human Molecular Genetics Area—Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain
- Correspondence: (A.V.); (M.W.B.)
| | - Martin W. Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen, Denmark
- Correspondence: (A.V.); (M.W.B.)
| |
Collapse
|
3
|
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
|
4
|
Nguyen NT, Han W, Cao W, Wang Y, Wen S, Huang Y, Li M, Du L, Zhou Y. Store‐Operated Calcium Entry Mediated by ORAI and STIM. Compr Physiol 2018; 8:981-1002. [DOI: 10.1002/cphy.c170031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
5
|
Wang Y, Wang H, Li L, Li J, Pan T, Zhang D, Yang H. Elevated expression of STIM1 is involved in lung tumorigenesis. Oncotarget 2018; 7:86584-86593. [PMID: 27863410 PMCID: PMC5349937 DOI: 10.18632/oncotarget.13359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/29/2016] [Indexed: 12/11/2022] Open
Abstract
This study aimed to address the potential role of STIM1 (stromal interaction molecule 1) in lung tumorigenesis. Colony formation in soft agar assay and tumorigenicity in nude mice assay were conducted. Western blot, immunohistochemistry and quantitative real-time polymerase chain reaction were used to measure the STIM1 expression. The distribution of cell cycle was detected by flow cytometry assay. Our results showed that the expression of STIM1 mRNA was significantly higher in human lung tumors than that in adjacent non-neoplastic lung tissues. Significantly increased expression of STIM1 mRNA and protein was observed in 16HBE-benzo(a)pyrene (BaP) cells and in BaP-treated mice lung tissues compared with 16HBE-control cells and the control group, respectively. Silencing STIM1 inhibited the proliferation and colony formation of A549 cells in in vitro experiments, attenuated the growth of tumor xenografts of A549 cells in in vivo experiments and induced the arrest of cell cycle in the G1 phase. The markedly decreased expression of cyclin D1 protein was observed in A549-shRNA-STIM1 cells as compared to A549-shRNA-control cells. The markedly increased expression of p21 protein was observed in A549-shRNA-STIM1 cells as compared to A549-shRNA-control cells. The expression levels of β-catenin and TGIF proteins were lower in A549-shRNA-STIM1 cells than those in A549-shRNA-control cells. In conclusion, this study indicated that the elevated expression of STIM1 might be involved in lung tumorigenesis.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Teng Pan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Ding Zhang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
6
|
Regulation of membrane ruffling by polarized STIM1 and ORAI1 in cortactin-rich domains. Sci Rep 2017; 7:383. [PMID: 28341841 PMCID: PMC5428229 DOI: 10.1038/s41598-017-00331-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/20/2017] [Indexed: 12/15/2022] Open
Abstract
Cell motility and migration requires the reorganization of the cortical cytoskeleton at the leading edge of cells and extracellular Ca2+ entry is essential for this reorganization. However the molecular nature of the regulators of this pathway is unknown. This work contributes to understanding the role of STIM1 and ORAI1 in the promotion of membrane ruffling by showing that phospho-STIM1 localizes at the leading edge of cells, and that both phospho-STIM1 and ORAI1 co-localize with cortactin (CTTN), a regulator of the cytoskeleton at membrane ruffling areas. STIM1-KO and ORAI1-KO cell lines were generated by CRISPR/Cas9 genome editing in U2OS cells. In both cases, KO cells presented a notable reduction of store-operated Ca2+ entry (SOCE) that was rescued by expression of STIM1-mCherry and ORAI1-mCherry. These results demonstrated that SOCE regulates membrane ruffling at the leading edge of cells. Moreover, endogenous ORAI1 and overexpressed ORAI1-GFP co-immunoprecipitated with endogenous CTTN. This latter result, in addition to the KO cells’ phenotype, the preservation of ORAI1-CTTN co-localization during ruffling, and the inhibition of membrane ruffling by the Ca2+-channel inhibitor SKF96365, further supports a functional link between SOCE and membrane ruffling.
Collapse
|
7
|
Barańska J, Czajkowski R, Pomorski P. P2Y 1 Receptors - Properties and Functional Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28639247 DOI: 10.1007/5584_2017_57] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this chapter we try to show a comprehensive image of current knowledge of structure, activity and physiological role of the P2Y1 purinergic receptor. The structure, distribution and changes in the expression of this receptor are summarized, as well as the mechanism of its signaling activity by the intracellular calcium mobilization. We try to show the connection between the components of its G protein activation and cellular or physiological effects, starting from changes in protein phosphorylation patterns and ending with such remote effects as receptor-mediated apoptosis. The special emphasis is put on the role of the P2Y1 receptor in cancer cells and neuronal plasticity. We concentrate on the P2Y1 receptor, it is though impossible to completely abstract from other aspects of nucleotide signaling and cross-talk with other nucleotide receptors is here discussed. Especially, the balance between P2Y1 and P2Y12 receptors, sharing the same ligand but signaling through different pathways, is presented.
Collapse
Affiliation(s)
- Jolanta Barańska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland
| | - Rafał Czajkowski
- Laboratory of Spatial Memory, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland
| | - Paweł Pomorski
- Laboratory of Molecular Basis of Cell Motility, Department of Cell Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland.
| |
Collapse
|
8
|
Jardin I, Rosado JA. STIM and calcium channel complexes in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1418-26. [DOI: 10.1016/j.bbamcr.2015.10.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/25/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
|
9
|
Chen YF, Hsu KF, Shen MR. The store-operated Ca 2+ entry-mediated signaling is important for cancer spread. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1427-35. [DOI: 10.1016/j.bbamcr.2015.11.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/17/2015] [Accepted: 11/25/2015] [Indexed: 12/31/2022]
|
10
|
Masuoka T, Kudo M, Yoshida J, Ishibashi T, Muramatsu I, Kato N, Imaizumi N, Nishio M. Long-Term Activation of Group I Metabotropic Glutamate Receptors Increases Functional TRPV1-Expressing Neurons in Mouse Dorsal Root Ganglia. Front Cell Neurosci 2016; 10:79. [PMID: 27064319 PMCID: PMC4814719 DOI: 10.3389/fncel.2016.00079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 12/17/2022] Open
Abstract
Damaged tissues release glutamate and other chemical mediators for several hours. These chemical mediators contribute to modulation of pruritus and pain. Herein, we investigated the effects of long-term activation of excitatory glutamate receptors on functional expression of transient receptor potential vaniloid type 1 (TRPV1) in dorsal root ganglion (DRG) neurons and then on thermal pain behavior. In order to detect the TRPV1-mediated responses in cultured DRG neurons, we monitored intracellular calcium responses to capsaicin, a TRPV1 agonist, with Fura-2. Long-term (4 h) treatment with glutamate receptor agonists (glutamate, quisqualate or DHPG) increased the proportion of neurons responding to capsaicin through activation of metabotropic glutamate receptor mGluR1, and only partially through the activation of mGluR5; engagement of these receptors was evident in neurons responding to allylisothiocyanate (AITC), a transient receptor potential ankyrin type 1 (TRPA1) agonist. Increase in the proportion was suppressed by phospholipase C (PLC), protein kinase C, mitogen/extracellular signal-regulated kinase, p38 mitogen-activated protein kinase or transcription inhibitors. Whole-cell recording was performed to record TRPV1-mediated membrane current; TRPV1 current density significantly increased in the AITC-sensitive neurons after the quisqualate treatment. To elucidate the physiological significance of this phenomenon, a hot plate test was performed. Intraplantar injection of quisqualate or DHPG induced heat hyperalgesia that lasted for 4 h post injection. This chronic hyperalgesia was attenuated by treatment with either mGluR1 or mGluR5 antagonists. These results suggest that long-term activation of mGluR1/5 by peripherally released glutamate may increase the number of neurons expressing functional TRPV1 in DRG, which may be strongly associated with chronic hyperalgesia.
Collapse
Affiliation(s)
- Takayoshi Masuoka
- Department of Pharmacology, School of Medicine, Kanazawa Medical University Uchinada, Japan
| | - Makiko Kudo
- Department of Pharmacology, School of Medicine, Kanazawa Medical University Uchinada, Japan
| | - Junko Yoshida
- Department of Pharmacology, School of Medicine, Kanazawa Medical University Uchinada, Japan
| | - Takaharu Ishibashi
- Department of Pharmacology, School of Medicine, Kanazawa Medical UniversityUchinada, Japan; Department of Pharmacology, School of Nursing, Kanazawa Medical UniversityUchinada, Japan
| | - Ikunobu Muramatsu
- Department of Pharmacology, School of Medicine, Kanazawa Medical University Uchinada, Japan
| | - Nobuo Kato
- Department of Physiology I, School of Medicine, Kanazawa Medical University Uchinada, Japan
| | - Noriko Imaizumi
- Department of Pharmacology, School of Medicine, Kanazawa Medical University Uchinada, Japan
| | - Matomo Nishio
- Department of Pharmacology, School of Medicine, Kanazawa Medical University Uchinada, Japan
| |
Collapse
|
11
|
Cheng H, Wang S, Feng R. STIM1 plays an important role in TGF-β-induced suppression of breast cancer cell proliferation. Oncotarget 2016; 7:16866-78. [PMID: 26919241 PMCID: PMC4941356 DOI: 10.18632/oncotarget.7619] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/11/2016] [Indexed: 01/08/2023] Open
Abstract
Store-operated calcium entry (SOCE) signaling is involved in cancer progression. Stromal interaction molecule 1 (STIM1) triggers store-operated calcium channels to induce SOCE. Transforming growth factor-β (TGF-β) influences a wide range of cellular behaviors, including cell proliferation. However, little is known about the relationship between calcium signaling and TGF-β signaling in cancer cell proliferation. Here, we found that TGF-β induced cell cycle arrest at the G0/G1 phase and suppressed cell proliferation in MDA-MB-231 and MCF-7 breast cancer cells. These effects were impaired by extracellular Ca2+ chelator EGTA or SOCE specific inhibitor SKF96365 in MDA-MB-231 cells. Treating MDA-MB-231 cells with TGF-β for 24 and 48 h markedly decreased STIM1 expression and thapsigargin-induced SOCE. A transcriptional inhibitor of STIM1, Wilm's tumor suppressor 1 (WT1), was upregulated in TGF-β-treated MDA-MB-231 cells, and knockdown of WT1 expression partially restored the TGF-β-induced downregulation of STIM1. Stably overexpressing STIM1 in MDA-MB-231 cells restored the TGF-β-induced effects. The p21 mRNA level increased in SKF96365- or TGF-β-treated MDA-MB-231 cells, whereas that for cyclin E1 decreased. Our findings demonstrate for the first time that STIM1 and SOCE are involved in the TGF-β-induced suppression of cell proliferation. Furthermore, our studies also provide a new approach to inhibit breast cancer cell proliferation with small molecules targeting STIM1 and SOCE.
Collapse
Affiliation(s)
- Huanyi Cheng
- State Key Laboratory of Membrane Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Shiqiang Wang
- State Key Laboratory of Membrane Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Renqing Feng
- State Key Laboratory of Membrane Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
12
|
Hoth M. CRAC channels, calcium, and cancer in light of the driver and passenger concept. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1408-17. [PMID: 26705695 DOI: 10.1016/j.bbamcr.2015.12.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 01/18/2023]
Abstract
Advances in next-generation sequencing allow very comprehensive analyses of large numbers of cancer genomes leading to an increasingly better characterization and classification of cancers. Comparing genomic data predicts candidate genes driving development, growth, or metastasis of cancer. Cancer driver genes are defined as genes whose mutations are causally implicated in oncogenesis whereas passenger mutations are defined as not being oncogenic. Currently, a list of several hundred cancer driver mutations is discussed including prominent members like TP53, BRAF, NRAS, or NF1. According to the vast literature on Ca(2+) and cancer, Ca(2+) signals and the underlying Ca(2+) channels and transporters certainly influence the development, growth, and metastasis of many cancers. In this review, I focus on the calcium release-activated calcium (CRAC) channel genes STIM and Orai and their role for cancer development, growth, and metastasis. STIM and Orai genes are being discussed in the context of current cancer concepts with a focus on the driver-passenger hypothesis. One result of this discussion is the hypothesis that a driver analysis of Ca(2+) homeostasis-related genes should not be carried out by looking at isolated genes. Rather a pool of “Ca(2+) genes” might be considered to act as one potential cancer driver. This article is part of a Special Issue entitled: Calcium and Cell Fate. Guest Editors: Jacques Haiech, Claus Heizmann, Joachim Krebs, Thierry Capiod and Olivier Mignen.
Collapse
Affiliation(s)
- Markus Hoth
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Building 48, Saarland University, D-66421 Homburg, Germany.
| |
Collapse
|
13
|
Xie J, Pan H, Yao J, Zhou Y, Han W. SOCE and cancer: Recent progress and new perspectives. Int J Cancer 2015; 138:2067-77. [PMID: 26355642 PMCID: PMC4764496 DOI: 10.1002/ijc.29840] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 09/03/2015] [Indexed: 12/15/2022]
Abstract
Ca2+ acts as a universal and versatile second messenger in the regulation of a myriad of biological processes, including cell proliferation, differentiation, migration and apoptosis. Store‐operated Ca2+ entry (SOCE) mediated by ORAI and the stromal interaction molecule (STIM) constitutes one of the major routes of calcium entry in nonexcitable cells, in which the depletion of intracellular Ca2+ stores triggers activation of the endoplasmic reticulum (ER)‐resident Ca2+ sensor protein STIM to gate and open the ORAI Ca2+ channels in the plasma membrane (PM). Accumulating evidence indicates that SOCE plays critical roles in cancer cell proliferation, metastasis and tumor neovascularization, as well as in antitumor immunity. We summarize herein the recent advances in our understanding of the function of SOCE in various types of tumor cells, vascular endothelial cells and cells of the immune system. Finally, the therapeutic potential of SOCE inhibitors in the treatment of cancer is also discussed.
Collapse
Affiliation(s)
- Jiansheng Xie
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongming Pan
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junlin Yao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX
| | - Weidong Han
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Wang JY, Sun J, Huang MY, Wang YS, Hou MF, Sun Y, He H, Krishna N, Chiu SJ, Lin S, Yang S, Chang WC. STIM1 overexpression promotes colorectal cancer progression, cell motility and COX-2 expression. Oncogene 2015; 34:4358-67. [PMID: 25381814 PMCID: PMC4426254 DOI: 10.1038/onc.2014.366] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 09/11/2014] [Accepted: 09/19/2014] [Indexed: 12/16/2022]
Abstract
Tumor metastasis is the major cause of death among cancer patients, with >90% of cancer-related death attributable to the spreading of metastatic cells to secondary organs. Store-operated Ca(2+) entry (SOCE) is the predominant Ca(2+) entry mechanism in most cancer cells, and stromal interaction molecule 1 (STIM1) is the endoplasmic reticulum (ER) Ca(2+) sensor for store-operated channels. Here we reported that the STIM1 was overexpressed in colorectal cancer (CRC) patients. STIM1 overexpression in CRC was significantly associated with tumor size, depth of invasion, lymph node metastasis status and serum levels of carcinoembryonic antigen. Furthermore, ectopic expression of STIM1 promoted CRC cell motility, while depletion of STIM1 with short hairpin RNA inhibited CRC cell migration. Our data further suggested that STIM1 promoted CRC cell migration through increasing the expression of cyclooxygenase-2 (COX-2) and production of prostaglandin E2 (PGE2). Importantly, ectopically expressed COX-2 or exogenous PGE2 were able to rescue migration defect in STIM1 knockdown CRC cells, and inhibition of COX-2 with ibuprofen and indomethacin abrogated STIM1-mediated CRC cell motility. In short, our data provided clinicopathological significance for STIM1 and SOCE in CRC progression, and implicated a role for COX-2 in STIM1-mediated CRC metastasis. Our studies also suggested a new approach to inhibit STIM1-mediated metastasis with COX-2 inhibitors.
Collapse
Affiliation(s)
- Jaw-Yuan Wang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Division of Gastrointestinal and General Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jianwei Sun
- Comprehensive Melanoma Research Center, Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Ming-Yii Huang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yu-Shiuan Wang
- Department of Genomic Medicine, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Surgery, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Yan Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Huifang He
- Comprehensive Melanoma Research Center, Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Niveditha Krishna
- Comprehensive Melanoma Research Center, Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Siou-Jin Chiu
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Shengchen Lin
- Comprehensive Melanoma Research Center, Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Shengyu Yang
- Comprehensive Melanoma Research Center, Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Wei-Chiao Chang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Clinical Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
15
|
Xu Y, Zhang S, Niu H, Ye Y, Hu F, Chen S, Li X, Luo X, Jiang S, Liu Y, Chen Y, Li J, Xiang R, Li N. STIM1 accelerates cell senescence in a remodeled microenvironment but enhances the epithelial-to-mesenchymal transition in prostate cancer. Sci Rep 2015; 5:11754. [PMID: 26257076 PMCID: PMC4530453 DOI: 10.1038/srep11754] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
The importance of store-operated Ca2+ entry (SOCE) and the role of its key molecular regulators, STIM1 and ORAI1, in the development of cancer are emerging. Here, we report an unexpected dual function of SOCE in prostate cancer progression by revealing a decrease in the expression of STIM1 in human hyperplasia and tumor tissues of high histological grade and by demonstrating that STIM1 and ORAI1 inhibit cell growth by arresting the G0/G1 phase and enhancing cell senescence in human prostate cancer cells. In addition, STIM1 and ORAI1 inhibited NF-κB signaling and remodeled the tumor microenvironment by reducing the formation of M2 phenotype macrophages, possibly creating an unfavorable tumor microenvironment and inhibiting cancer development. However, STIM1 also promoted cell migration and the epithelial-to-mesenchymal transition by activating TGF-β, Snail and Wnt/β-Catenin pathways. Thus, our study revealed novel regulatory effects and the mechanisms by which STIM1 affects cell senescence, tumor migration and the tumor microenvironment, revealing that STIM1 has multiple functions in prostate cancer cells.
Collapse
Affiliation(s)
- Yingxi Xu
- 1] School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China [2] State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Shu Zhang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Haiying Niu
- Department of Obstetrics and Gynecology, First Central Hospital Clinic Institute, Tianjin Medical University, 24 Fukang Road, Tianjin 300192 China
| | - Yujie Ye
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Fen Hu
- School of Physics, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Si Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xuefei Li
- Beijing Health Vocational College, 94 Nanhengxijie Street, Beijing, 100053 China
| | - Xiaohe Luo
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shan Jiang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanhua Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanan Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Junying Li
- Department of Obstetrics and Gynecology, First Central Hospital Clinic Institute, Tianjin Medical University, 24 Fukang Road, Tianjin 300192 China
| | - Rong Xiang
- 1] School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China [2] Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China [3] Collaborative Innovation Center for Biotherapy, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Na Li
- 1] School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China [2] Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Tianjin 300071, China [3] Collaborative Innovation Center for Biotherapy, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
16
|
Vashisht A, Trebak M, Motiani RK. STIM and Orai proteins as novel targets for cancer therapy. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am J Physiol Cell Physiol 2015; 309:C457-69. [PMID: 26017146 DOI: 10.1152/ajpcell.00064.2015] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Calcium (Ca(2+)) regulates a plethora of cellular functions including hallmarks of cancer development such as cell cycle progression and cellular migration. Receptor-regulated calcium rise in nonexcitable cells occurs through store-dependent as well as store-independent Ca(2+) entry pathways. Stromal interaction molecules (STIM) and Orai proteins have been identified as critical constituents of both these Ca(2+) influx pathways. STIMs and Orais have emerged as targets for cancer therapeutics as their altered expression and function have been shown to contribute to tumorigenesis. Recent data demonstrate that they play a vital role in development and metastasis of a variety of tumor types including breast, prostate, cervical, colorectal, brain, and skin tumors. In this review, we will retrospect the data supporting a key role for STIM1, STIM2, Orai1, and Orai3 proteins in tumorigenesis and discuss the potential of targeting these proteins for cancer therapy.
Collapse
Affiliation(s)
- Ayushi Vashisht
- Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; and
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University School of Medicine, Hershey, Pennsylvania
| | - Rajender K Motiani
- Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; and
| |
Collapse
|
17
|
Ca2+ signaling in cytoskeletal reorganization, cell migration, and cancer metastasis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:409245. [PMID: 25977921 PMCID: PMC4421034 DOI: 10.1155/2015/409245] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/12/2015] [Indexed: 01/19/2023]
Abstract
Proper control of Ca2+ signaling is mandatory for effective cell migration, which is critical for embryonic development, wound healing, and cancer metastasis. However, how Ca2+ coordinates structural components and signaling molecules for proper cell motility had remained elusive. With the advance of fluorescent live-cell Ca2+ imaging in recent years, we gradually understand how Ca2+ is regulated spatially and temporally in migrating cells, driving polarization, protrusion, retraction, and adhesion at the right place and right time. Here we give an overview about how cells create local Ca2+ pulses near the leading edge, maintain cytosolic Ca2+ gradient from back to front, and restore Ca2+ depletion for persistent cell motility. Differential roles of Ca2+ in regulating various effectors and the interaction of roles of Ca2+ signaling with other pathways during migration are also discussed. Such information might suggest a new direction to control cancer metastasis by manipulating Ca2+ and its associating signaling molecules in a judicious manner.
Collapse
|
18
|
Wang X, Liu J, Jin NA, Xu D, Wang J, Han Y, Yin N. Fructus Corni extract-induced neuritogenesis in PC12 cells is associated with the suppression of stromal interaction molecule 1 expression and inhibition of Ca 2+ influx. Exp Ther Med 2015; 9:1773-1779. [PMID: 26136892 DOI: 10.3892/etm.2015.2316] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 02/13/2015] [Indexed: 12/19/2022] Open
Abstract
Fructus Corni (Cornus officinalis Sieb. et Zucc.) is commonly prescribed as a traditional Chinese herbal medicine that possesses pharmacological actions against inflammation, diabetic nephropathy, tumors, oxidation and aging. However, its function and mode of action within the nervous system remain largely unclear. In this study, the effects of Fructus Corni extract (FCE) on neuronal differentiation were investigated. It was found that FCE significantly increased the percentage of PC12 cells bearing neurites (P<0.001). Following the generation of neurite outgrowth, FCE treatment decreased the mRNA expression of stromal interaction molecule 1 (STIM1; P<0.05) and suppressed the expression of STIM1 protein (P<0.001). In addition, extracellular calcium (Ca2+) influx was inhibited resulting in a reduction in the intracellular Ca2+ level, suggesting that the inhibition of Ca2+ influx may be involved in the FCE-promoted neurite outgrowth of PC12 cells. These results demonstrate that FCE induces neurite outgrowth in PC12 cells and that this is associated with the suppression of STIM1 expression and the inhibition of Ca2+ influx, which may partially explain the FCE-induced neuritogenesis.
Collapse
Affiliation(s)
- Xushi Wang
- Department of Oncology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - Jiaqi Liu
- Department of Oncology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - N A Jin
- Department of Oncology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - Dan Xu
- Department of Oncology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - Junyu Wang
- Department of Oncology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - Yongming Han
- Department of Anatomy, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, P.R. China
| | - Nina Yin
- Department of Anatomy, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, P.R. China
| |
Collapse
|
19
|
STIM1 phosphorylation triggered by epidermal growth factor mediates cell migration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:233-43. [PMID: 25447552 DOI: 10.1016/j.bbamcr.2014.10.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 11/23/2022]
Abstract
STIM1 is a key regulator of store-operated calcium entry (SOCE), and therefore a mediator of Ca²⁺ entry-dependent cellular events. Phosphorylation of STIM1 at ERK1/2 target sites has been described as enhancing STIM1 activation during intracellular Ca²⁺ emptying triggered by the inhibition of the sarco(endo)plasmic Ca²⁺ -ATPase with thapsigargin. However, no physiological function is known for this specific phosphorylation. The present study examined the role of STIM1 phosphorylation in cell signaling triggered by EGF. Using a human endometrial adenocarcinoma cell line (Ishikawa cells) EGF or H-Ras(G12V), an active mutant of H-Ras, was found to trigger STIM1 phosphorylation at residues Ser575, Ser608, and Ser621, and this process was sensitive to PD0325901, an inhibitor of ERK1/2. Both, ERK1/2 activation and STIM1 phosphorylation took place in the absence of extracellular Ca²⁺, indicating that both events are upstream steps for Ca²⁺entry activation. Also, EGF triggered the dissociation of STIM1 from EB1 (a regulator of microtubule plus-ends) in a manner similar to that reported for the activation of STIM1 by thapsigargin. Migration of the Ishikawa cells was impaired when STIM1 phosphorylation was targeted by Ser-to-Ala substitution mutation of ERK1/2 target sites. This effect was also observed with the Ca²⁺ channel blocker SKF96365. Phosphomimetic mutation of STIM1 restored the migration to levels similar to that found for STIM1-wild type. Finally, the increased vimentin expression and relocalization of E-cadherin triggered by EGF were largely inhibited by targeting STIM1 phosphorylation, while STIM1-S575E/S608E/S621E normalized the profiles of these two EMT markers.
Collapse
|
20
|
Sobradillo D, Hernández-Morales M, Ubierna D, Moyer MP, Núñez L, Villalobos C. A reciprocal shift in transient receptor potential channel 1 (TRPC1) and stromal interaction molecule 2 (STIM2) contributes to Ca2+ remodeling and cancer hallmarks in colorectal carcinoma cells. J Biol Chem 2014; 289:28765-82. [PMID: 25143380 DOI: 10.1074/jbc.m114.581678] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We have investigated the molecular basis of intracellular Ca(2+) handling in human colon carcinoma cells (HT29) versus normal human mucosa cells (NCM460) and its contribution to cancer features. We found that Ca(2+) stores in colon carcinoma cells are partially depleted relative to normal cells. However, resting Ca(2+) levels, agonist-induced Ca(2+) increases, store-operated Ca(2+) entry (SOCE), and store-operated currents (ISOC) are largely enhanced in tumor cells. Enhanced SOCE and depleted Ca(2+) stores correlate with increased cell proliferation, invasion, and survival characteristic of tumor cells. Normal mucosa cells displayed small, inward Ca(2+) release-activated Ca(2+) currents (ICRAC) mediated by ORAI1. In contrast, colon carcinoma cells showed mixed currents composed of enhanced ICRAC plus a nonselective ISOC mediated by TRPC1. Tumor cells display increased expression of TRPC1, ORAI1, ORAI2, ORAI3, and STIM1. In contrast, STIM2 protein was nearly depleted in tumor cells. Silencing data suggest that enhanced ORAI1 and TRPC1 contribute to enhanced SOCE and differential store-operated currents in tumor cells, whereas ORAI2 and -3 are seemingly less important. In addition, STIM2 knockdown decreases SOCE and Ca(2+) store content in normal cells while promoting apoptosis resistance. These data suggest that loss of STIM2 may underlie Ca(2+) store depletion and apoptosis resistance in tumor cells. We conclude that a reciprocal shift in TRPC1 and STIM2 contributes to Ca(2+) remodeling and tumor features in colon cancer.
Collapse
Affiliation(s)
- Diego Sobradillo
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Miriam Hernández-Morales
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Daniel Ubierna
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | | | - Lucía Núñez
- the Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, 47003 Valladolid, Spain
| | - Carlos Villalobos
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain,
| |
Collapse
|
21
|
Pierro C, Cook SJ, Foets TCF, Bootman MD, Roderick HL. Oncogenic K-Ras suppresses IP₃-dependent Ca²⁺ release through remodelling of the isoform composition of IP₃Rs and ER luminal Ca²⁺ levels in colorectal cancer cell lines. J Cell Sci 2014; 127:1607-19. [PMID: 24522186 DOI: 10.1242/jcs.141408] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The GTPase Ras is a molecular switch engaged downstream of G-protein-coupled receptors and receptor tyrosine kinases that controls multiple cell-fate-determining signalling pathways. Ras signalling is frequently deregulated in cancer, underlying associated changes in cell phenotype. Although Ca(2+) signalling pathways control some overlapping functions with Ras, and altered Ca(2+) signalling pathways are emerging as important players in oncogenic transformation, how Ca(2+) signalling is remodelled during transformation and whether it has a causal role remains unclear. We have investigated Ca(2+) signalling in two human colorectal cancer cell lines and their isogenic derivatives in which the allele encoding oncogenic K-Ras (G13D) was deleted by homologous recombination. We show that agonist-induced Ca(2+) release from the endoplasmic reticulum (ER) intracellular Ca(2+) stores is enhanced by loss of K-Ras(G13D) through an increase in the Ca(2+) content of the ER store and a modification of the abundance of inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) subtypes. Consistently, uptake of Ca(2+) into mitochondria and sensitivity to apoptosis was enhanced as a result of K-Ras(G13D) loss. These results suggest that suppression of Ca(2+) signalling is a common response to naturally occurring levels of K-Ras(G13D), and that this contributes to a survival advantage during oncogenic transformation.
Collapse
Affiliation(s)
- Cristina Pierro
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT UK
| | | | | | | | | |
Collapse
|
22
|
Berchtold MW, Villalobo A. The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:398-435. [PMID: 24188867 DOI: 10.1016/j.bbamcr.2013.10.021] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/24/2013] [Accepted: 10/26/2013] [Indexed: 12/21/2022]
Abstract
Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.
Collapse
Key Words
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-ethyl]-4,5-dihydro-pyrazol-1-yl]-benzoic acid
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-vinyl]-4,5-dihydro-pyrazol-1-yl]-phenyl)-(4-methyl-piperazin-1-yl)-methanone
- (−) enantiomer of dihydropyrine 3-methyl-5-3-(4,4-diphenyl-1-piperidinyl)-propyl-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-piridine-3,5-dicarboxylate-hydrochloride (niguldipine)
- 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine
- 12-O-tetradecanoyl-phorbol-13-acetate
- 2-chloro-(ε-amino-Lys(75))-[6-(4-(N,N′-diethylaminophenyl)-1,3,5-triazin-4-yl]-CaM adduct
- 3′-(β-chloroethyl)-2′,4′-dioxo-3,5′-spiro-oxazolidino-4-deacetoxy-vinblastine
- 7,12-dimethylbenz[a]anthracene
- Apoptosis
- Autophagy
- B859-35
- CAPP(1)-CaM
- Ca(2+) binding protein
- Calmodulin
- Cancer biology
- Cell proliferation
- DMBA
- EBB
- FL-CaM
- FPCE
- HBC
- HBCP
- J-8
- KAR-2
- KN-62
- KN-93
- N-(4-aminobutyl)-2-naphthalenesulfonamide
- N-(4-aminobutyl)-5-chloro-2-naphthalenesulfonamide
- N-(6-aminohexyl)-1-naphthalenesulfonamide
- N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide
- N-8-aminooctyl-5-iodo-naphthalenesulfonamide
- N-[2-[N-(4-chlorocinnamyl)-N-methylaminomethyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide
- O-(4-ethoxyl-butyl)-berbamine
- RITC-CaM
- TA-CaM
- TFP
- TPA
- W-12
- W-13
- W-5
- W-7
- fluorescein-CaM adduct
- fluphenazine-N-2-chloroethane
- norchlorpromazine-CaM adduct
- rhodamine isothiocyanate-CaM adduct
- trifluoperazine
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, Copenhagen Biocenter 4-2-09 Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Department of Cancer Biology, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
23
|
Activation of the endoplasmic reticulum calcium sensor STIM1 and store-operated calcium entry by rotavirus requires NSP4 viroporin activity. J Virol 2013; 87:13579-88. [PMID: 24109210 DOI: 10.1128/jvi.02629-13] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Rotavirus nonstructural protein 4 (NSP4) induces dramatic changes in cellular calcium homeostasis. These include increased endoplasmic reticulum (ER) permeability, resulting in decreased ER calcium stores and activation of plasma membrane (PM) calcium influx channels, ultimately causing a 2- to 4-fold elevation in cytoplasmic calcium. Elevated cytoplasmic calcium is absolutely required for virus replication, but the underlying mechanisms responsible for calcium influx remain poorly understood. NSP4 is an ER-localized viroporin, whose activity depletes ER calcium, which ultimately leads to calcium influx. We hypothesized that NSP4-mediated depletion of ER calcium activates store-operated calcium entry (SOCE) through activation of the ER calcium sensor stromal interaction molecule 1 (STIM1). We established and used a stable yellow fluorescent protein-expressing STIM1 cell line (YFP-STIM1) as a biosensor to assess STIM1 activation (puncta formation) by rotavirus infection and NSP4 expression. We found that STIM1 is constitutively active in rotavirus-infected cells and that STIM1 puncta colocalize with the PM-localized Orai1 SOCE calcium channel. Expression of wild-type NSP4 activated STIM1, resulting in PM calcium influx, but an NSP4 viroporin mutant failed to induce STIM1 activation and did not activate the PM calcium entry pathway. Finally, knockdown of STIM1 significantly reduced rotavirus yield, indicating STIM1 plays a critical role in virus replication. These data demonstrate that while rotavirus may ultimately activate multiple calcium channels in the PM, calcium influx is predicated on NSP4 viroporin-mediated activation of STIM1 in the ER. This is the first report of viroporin-mediated activation of SOCE, reinforcing NSP4 as a robust model to understand dysregulation of calcium homeostasis during virus infections.
Collapse
|
24
|
Casas-Rua V, Alvarez IS, Pozo-Guisado E, Martín-Romero FJ. Inhibition of STIM1 phosphorylation underlies resveratrol-induced inhibition of store-operated calcium entry. Biochem Pharmacol 2013; 86:1555-63. [PMID: 24095720 DOI: 10.1016/j.bcp.2013.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 11/19/2022]
Abstract
Resveratrol, a natural phytoalexin that shows health-promoting benefits, is an inhibitor of store-operated calcium entry (SOCE). Knowledge of the molecular mechanism underlying this inhibition is required for the proper design of therapies that include resveratrol or related stilbenoids, but remains largely unknown. To unravel this mechanism, using HEK293 cells as a model, we found that resveratrol inhibited the ERK1/2 activation triggered by Ca²⁺ store depletion. As a consequence, resveratrol inhibited STIM1 phosphorylation at residues Ser575, Ser608, and Ser621. Because this phosphorylation regulates the dissociation of STIM1 from the microtubule plus-end binding protein EB1 under store depletion conditions, resveratrol inhibited STIM1-EB1 dissociation. This inhibition had downstream effects such as inhibition of STIM1 multimerization in response to store depletion, and a significant impairment in the binding of STIM1 to ORAI1. Although additional targets for resveratrol in the molecular mechanism that governs SOCE cannot be discarded, the present results demonstrate that ERK1/2 pathway is a major target for resveratrol, and that the impairment of its activation produces a significant inhibition of SOCE.
Collapse
Affiliation(s)
- Vanessa Casas-Rua
- Department of Biochemistry and Molecular Biology, School of Life Sciences, University of Extremadura, Badajoz, Spain; Department of Cell Biology, School of Life Sciences, University of Extremadura, Badajoz, Spain.
| | | | | | | |
Collapse
|
25
|
Soltoff SP, Lannon WA. Activation of ERK1/2 by store-operated calcium entry in rat parotid acinar cells. PLoS One 2013; 8:e72881. [PMID: 24009711 PMCID: PMC3756958 DOI: 10.1371/journal.pone.0072881] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 07/15/2013] [Indexed: 12/23/2022] Open
Abstract
The regulation of intracellular Ca2+ concentration ([Ca2+]i) plays a critical role in a variety of cellular processes, including transcription, protein activation, vesicle trafficking, and ion movement across epithelial cells. In many cells, the activation of phospholipase C-coupled receptors hydrolyzes membrane phosphoinositides and produces the depletion of endoplasmic reticulum Ca2+ stores, followed by the sustained elevation of [Ca2+]i from Ca2+ entry across the plasma membrane via store-operated Ca2+ entry (SOCE). Ca2+ entry is also increased in a store-independent manner by arachidonate-regulated Ca2+ (ARC) channels. Using rat parotid salivary gland cells, we examined multiple pathways of Ca2+ entry/elevation to determine if they activated cell signaling proteins and whether this occurred in a pathway-dependent manner. We observed that SOCE activates extracellular signal-related kinases 1 and 2 (ERK1/2) to ∼3-times basal levels via a receptor-independent mechanism when SOCE was initiated by depleting Ca2+ stores using the endoplasmic reticulum Ca2+-ATPase inhibitor thapsigargin (TG). TG-initiated ERK1/2 phosphorylation increased as rapidly as that initiated by the muscarinic receptor agonist carbachol, which promoted an increase to ∼5-times basal levels. Notably, ERK1/2 phosphorylation was not increased by the global elevation of [Ca2+]i by Ca2+ ionophore or by Ca2+ entry via ARC channels in native cells, although ERK1/2 phosphorylation was increased by Ca2+ ionophore in Par-C10 and HSY salivary cell lines. Agents and conditions that blocked SOCE in native cells, including 2-aminoethyldiphenyl borate (2-APB), SKF96363, and removal of extracellular Ca2+, also reduced TG- and carbachol-stimulated ERK1/2 phosphorylation. TG-promoted ERK1/2 phosphorylation was blocked when SRC and Protein Kinases C (PKC) were inhibited, and it was blocked in cells pretreated with β-adrenergic agonist isoproterenol. These observations demonstrate that ERK1/2 is activated by a selective mechanism of Ca2+ entry (SOCE) in these cells, and suggest that ERK1/2 may contribute to events downstream of SOCE.
Collapse
Affiliation(s)
- Stephen P Soltoff
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Signal Transduction, Harvard Medical School, Boston, Massachussetts, USA.
| | | |
Collapse
|
26
|
Yang IH, Tsai YT, Chiu SJ, Liu LT, Lee HH, Hou MF, Hsu WL, Chen BK, Chang WC. Involvement of STIM1 and Orai1 in EGF-mediated cell growth in retinal pigment epithelial cells. J Biomed Sci 2013; 20:41. [PMID: 23800047 PMCID: PMC3700824 DOI: 10.1186/1423-0127-20-41] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/18/2013] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In non-excitable cells, one major route for calcium entry is through store-operated calcium (SOC) channels in the plasma membrane. These channels are activated by the emptying of intracellular Ca²⁺ store. STIM1 and Orai1 are major regulators of SOC channels. In this study, we explored the functions of STIM1 and Orai1 in epidermal growth factor (EGF)-induced cell proliferation and migration in retinal pigment epithelial cells (ARPE-19 cell line). RESULTS EGF triggers cell proliferation and migration in ARPE-19 cells. Cell proliferation and migration involve STIM1 and Orai1, as well as phosphorylation of extracellular signal-regulated protein kinase (ERK) 1/2, and Akt. Pharmacological inhibitors of SOC channels and siRNA of Orai1 and STIM1 suppress cell proliferation and migration. Pre-treatment of mitogen-activated protein kinase kinase (MEK) inhibitors and a phosphatidylinositol 3 kinases (PI3K) inhibitor attenuated cell proliferation and migration. However, inhibition of the SOC channels failed to prevent EGF-mediated ERK 1/2 and Akt phosphorylation. CONCLUSIONS Our results showed that STIM1, Orai1, ERK 1/2, and Akt are key determinants of EGF-mediated cell growth in ARPE-19 cells. EGF is a potent growth molecule that has been linked to the development of PVR, and therefore, STIM1, Orai1, as well as the MEK/ERK 1/2 and PI3K/Akt pathways, might be potential therapeutic targets for drugs aimed at treating such disorders.
Collapse
Affiliation(s)
- I-Hui Yang
- Department of Medical Genetics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|