101
|
Li S, Wang J, Wei Y, Liu Y, Ding X, Dong B, Xu Y, Wang Y. Critical role of TRPC6 in maintaining the stability of HIF-1α in glioma cells under hypoxia. J Cell Sci 2015; 128:3317-29. [PMID: 26187851 DOI: 10.1242/jcs.173161] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/07/2015] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a key transcriptional factor responsible for the expression of a broad range of genes that facilitate acclimatization to hypoxia. Its stability is predominantly controlled by rapid hydroxylation of two prolines on its α subunit. However, how the rapid hydroxylation of HIF-1α is regulated is not fully understood. Here, we report that transient receptor potential canonical (TRPC) 6 channels control hydroxylation and stability of HIF-1α in human glioma cells under hypoxia. TRPC6 was rapidly activated by IGF-1R-PLCγ-IP3R pathway in hypoxia. Inhibition of TRPC6 enhanced the levels of α-ketoglutarate (α-KG) and promoted hydroxylation of HIF-1α to suppress HIF-1α accumulation without affecting its transcription or translation. Dimethyloxalylglycine N-(methoxyoxoacetyl)-glycine methyl ester (DMOG), an analog of α-KG, reversed the inhibition of HIF-1α accumulation. Moreover, TRPC6 regulated GLUT1 expression depending on HIF-1α accumulation to affect glucose uptake in hypoxia. Our results suggest that TRPC6 regulates metabolism to affect HIF-1α stability and consequent glucose metabolism in human glioma cells under hypoxia.
Collapse
Affiliation(s)
- Shanshan Li
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinkui Wang
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yi Wei
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yongjian Liu
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xia Ding
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Dong
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yinghui Xu
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
102
|
Gautier M, Dhennin-Duthille I, Ay AS, Rybarczyk P, Korichneva I, Ouadid-Ahidouch H. New insights into pharmacological tools to TR(i)P cancer up. Br J Pharmacol 2014; 171:2582-92. [PMID: 24345078 DOI: 10.1111/bph.12561] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 12/09/2013] [Accepted: 12/13/2013] [Indexed: 12/25/2022] Open
Abstract
The aim of this review is to address the recent advances regarding the use of pharmacological agents to target transient receptor potential (TRP) channels in cancer and their potential application in therapeutics. Physiologically, TRP channels are responsible for cation entry (Ca(2+) , Na(+) , Mg(2+) ) in many mammalian cells and regulate a large number of cellular functions. However, dysfunction in channel expression and/or activity can be linked to human diseases like cancer. Indeed, there is growing evidence that TRP channel expression is altered in cancer tissues in comparison with normal ones. Moreover, these proteins are involved in many cancerous processes, including cell proliferation, apoptosis, migration and invasion, as well as resistance to chemotherapy. Among the TRP superfamily, TRPC, TRPV, TRPM and TRPA1 have been shown to play a role in many cancer types, including breast, digestive, gliomal, head and neck, lung and prostate cancers. Pharmacological modulators are used to characterize the functional implications of TRP channels in whole-cell membrane currents, resting membrane potential regulation and intracellular Ca(2+) signalling. Moreover, pharmacological modulation of TRP activity in cancer cells is systematically linked to the effect on cancerous processes (proliferation, survival, migration, invasion, sensitivity to chemotherapeutic drugs). Here we describe the effects of such TRP modulators on TRP activity and cancer cell phenotype. Furthermore, the potency and specificity of these agents will be discussed, as well as the development of new strategies for targeting TRP channels in cancer.
Collapse
Affiliation(s)
- M Gautier
- Laboratory of Cell and Molecular Physiology, SFR CAP-Santé (FED 4231), University of Picardie Jules Verne, UFR Sciences, EA 4667, Amiens, France
| | | | | | | | | | | |
Collapse
|
103
|
Wang T, Chen Z, Zhu Y, Pan Q, Liu Y, Qi X, Jin L, Jin J, Ma X, Hua D. Inhibition of transient receptor potential channel 5 reverses 5-Fluorouracil resistance in human colorectal cancer cells. J Biol Chem 2014; 290:448-56. [PMID: 25404731 DOI: 10.1074/jbc.m114.590364] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
5-Fluorouracil (5-Fu) is commonly used in the chemotherapy of colorectal cancer (CRC), but resistance to 5-Fu occurs in most cases, allowing cancer progression. Suppressing ABCB1 (ATP-binding cassette, subfamily B, member 1), which is a pump overproduced in cancer cells to export cytotoxic drugs, is an attractive strategy to overcome drug resistance. In the present study, transient receptor potential channel TrpC5 was found to be overproduced at the mRNA and protein levels together with ABCB1 in 5-Fu-resistant human CRC HCT-8 (HCT-8/5-Fu) and LoVo (LoVo/5-Fu) cells. More nuclear-stabilized β-catenin accumulation was found in HCT-8/5-Fu and LoVo/5-Fu cells than in HCT-8 and LoVo cells. Suppressing TrpC5 expression with TrpC5-specific siRNA inhibited the canonical Wnt/β-catenin signal pathway, reduced the induction of ABCB1, weakened the ABCB1 efflux pump, and caused a remarkable reversal of 5-Fu resistance in HCT-8/5-Fu and LoVo/5-Fu cells. On the contrary, enforcing TrpC5 expression resulted in an activated Wnt/β-catenin signal pathway and up-regulation of ABCB1. Taken together, we demonstrated an essential role of TrpC5 in ABCB1 induction and drug resistance in human CRC cells via promoting nuclear β-catenin accumulation.
Collapse
Affiliation(s)
- Teng Wang
- From the Departments of Medical Oncology and
| | - Zhen Chen
- the School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yifei Zhu
- the School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qiongxi Pan
- the School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanjun Liu
- the School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiaowei Qi
- Pathology, Affiliated Hospital of Jiangnan University and Fourth People's Hospital of Wuxi, Wuxi, Jiangsu 214062, China and
| | - Linfang Jin
- Pathology, Affiliated Hospital of Jiangnan University and Fourth People's Hospital of Wuxi, Wuxi, Jiangsu 214062, China and
| | - Jian Jin
- the School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xin Ma
- the School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Dong Hua
- From the Departments of Medical Oncology and
| |
Collapse
|
104
|
Chen J, Luan Y, Yu R, Zhang Z, Zhang J, Wang W. Transient receptor potential (TRP) channels, promising potential diagnostic and therapeutic tools for cancer. Biosci Trends 2014; 8:1-10. [PMID: 24647107 DOI: 10.5582/bst.8.1] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Despite the advances in detection of and therapies for various tumors, high rates of treatment failure and mortality still exist throughout the world. These high rates are mainly due to the powerful capability of tumor cells to proliferate and migrate. Recent studies regarding the transient receptor potential (TRP) have indicated that TRP channels are associated with tumors and that TRP channels might represent potential targets for cancer treatment. TRP channels are important calcium-selective ion channels in many different tissues and cell types in mammals and are crucial regulators of calcium and sodium. TRP were first discovered in the photoreceptors of Drosophila with gene defects or mutations. TRP channels can be divided into seven subfamilies: TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPML (mucolipin), TRPP (polycystin), TRPA (ankyrin transmembrane protein), and TRPN (NomPC-like). TRPC proteins are conserved across organisms since they are most homologous to Drosophila TRP. TRP superfamilies have been linked to many physiological and pathological functions, including cell differentiation, proliferation, apoptosis, and ion homeostasis. This review focuses on the properties of TRP in oncogenesis, cancer proliferation, and cell migration.
Collapse
Affiliation(s)
- Jianpeng Chen
- Department of Oncology, Provincial Hospital Affiliated with Shandong University
| | | | | | | | | | | |
Collapse
|
105
|
Huang L, Li B, Tang S, Guo H, Li W, Huang X, Yan W, Zou F. Mitochondrial KATP Channels Control Glioma Radioresistance by Regulating ROS-Induced ERK Activation. Mol Neurobiol 2014; 52:626-37. [PMID: 25249341 DOI: 10.1007/s12035-014-8888-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/29/2014] [Indexed: 12/01/2022]
Abstract
Malignant glioma is the most prevalent form of malignant brain tumor. Although radiotherapy is widely used in glioma treatment, the radioresistance of glioma cells limits the success of the glioma treatment. The lack of effective targets and signaling pathways to reverse glioma radioresistance is the critical obstacle in successful treatment. In this study, we demonstrate that mitochondrial ATP-sensitive potassium channels (mtK(ATP) channels) are overexpressed in glioma cells and are closely related to the malignancy grade and the overall survival of the patients. Importantly, we showed that mtK(ATP) channels could control glioma radioresistance by regulating reactive oxygen species (ROS)-induced ERK activation. The inhibition of mtK(ATP) channels suppresses glioma radioresistance by inhibiting ERK activation both in vitro and in vivo. These findings reveal the important roles of the mitochondria and mtK(ATP) channels as key regulators in the radioresistance of glioma cells, and suggest that mtK(ATP) channel blockers and MAPK/ERK kinase (MEK) inhibitors are potential targets for drug development of glioma treatments.
Collapse
Affiliation(s)
- Lianyan Huang
- Department of Occupational Health and Occupational Medicine, School of Public Health and Tropical Medicine, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | | | | | | | | | | | | | | |
Collapse
|
106
|
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: 91] [Impact Index Per Article: 8.3] [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
|
107
|
Stewart TA, Yapa KTDS, Monteith GR. Altered calcium signaling in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2502-11. [PMID: 25150047 DOI: 10.1016/j.bbamem.2014.08.016] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/11/2014] [Indexed: 01/03/2023]
Abstract
It is the nature of the calcium signal, as determined by the coordinated activity of a suite of calcium channels, pumps, exchangers and binding proteins that ultimately guides a cell's fate. Deregulation of the calcium signal is often deleterious and has been linked to each of the 'cancer hallmarks'. Despite this, we do not yet have a full understanding of the remodeling of the calcium signal associated with cancer. Such an understanding could aid in guiding the development of therapies specifically targeting altered calcium signaling in cancer cells during tumorigenic progression. Findings from some of the studies that have assessed the remodeling of the calcium signal associated with tumorigenesis and/or processes important in invasion and metastasis are presented in this review. The potential of new methodologies is also discussed. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Teneale A Stewart
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Kunsala T D S Yapa
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
108
|
Store-operated Ca2+ entry does not control proliferation in primary cultures of human metastatic renal cellular carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:739494. [PMID: 25126575 PMCID: PMC4119920 DOI: 10.1155/2014/739494] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/29/2014] [Accepted: 05/19/2014] [Indexed: 11/18/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is activated following depletion of the inositol-1,4,5-trisphosphate (InsP3)-sensitive Ca2+ pool to regulate proliferation in immortalized cell lines established from either primary or metastatic lesions. The molecular nature of SOCE may involve both Stim1, which senses Ca2+ levels within the endoplasmic reticulum (ER) Ca2+ reservoir, and a number of a Ca2+-permeable channels on the plasma membrane, including Orai1, Orai3, and members of the canonical transient receptor (TRPC1–7) family of ion channels. The present study was undertaken to assess whether SOCE is expressed and controls proliferation in primary cultures isolated from secondary lesions of heavily pretreated metastatic renal cell carcinoma (mRCC) patients. SOCE was induced following pharmacological depletion of the ER Ca2+ store, but not by InsP3-dependent Ca2+ release. Metastatic RCC cells express Stim1-2, Orai1–3, and TRPC1–7 transcripts and proteins. In these cells, SOCE was insensitive to BTP-2, 10 µM Gd3+ and Pyr6, while it was inhibited by 100 µM Gd3+, 2-APB, and carboxyamidotriazole (CAI). Neither Gd3+ nor 2-APB or CAI impaired mRCC cell proliferation. Consistently, no detectable Ca2+ signal was elicited by growth factor stimulation. Therefore, a functional SOCE is expressed but does not control proliferation of mRCC cells isolated from patients resistant to multikinase inhibitors.
Collapse
|
109
|
Nielsen N, Lindemann O, Schwab A. TRP channels and STIM/ORAI proteins: sensors and effectors of cancer and stroma cell migration. Br J Pharmacol 2014; 171:5524-40. [PMID: 24724725 DOI: 10.1111/bph.12721] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/24/2014] [Accepted: 04/03/2014] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Cancer cells are strongly influenced by host cells within the tumour stroma and vice versa. This leads to the development of a tumour microenvironment with distinct physical and chemical properties that are permissive for tumour progression. The ability to migrate plays a central role in this mutual interaction. Migration of cancer cells is considered as a prerequisite for tumour metastasis and the migration of host stromal cells is required for reaching the tumour site. Increasing evidence suggests that transient receptor potential (TRP) channels and STIM/ORAI proteins affect key calcium-dependent mechanisms implicated in both cancer and stroma cell migration. These include, among others, cytoskeletal remodelling, growth factor/cytokine signalling and production, and adaptation to tumour microenvironmental properties such as hypoxia and oxidative stress. In this review, we will summarize the current knowledge regarding TRP channels and STIM/ORAI proteins in cancer and stroma cell migration. We focus on how TRP channel or STIM/ORAI-mediated Ca(2+) signalling directly or indirectly influences cancer and stroma cell migration by affecting the above listed mechanisms. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- N Nielsen
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | | |
Collapse
|
110
|
Overexpression of transient receptor potential vanilloid 2 is associated with poor prognosis in patients with esophageal squamous cell carcinoma. Med Oncol 2014; 31:17. [PMID: 24878697 DOI: 10.1007/s12032-014-0017-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/28/2014] [Indexed: 12/23/2022]
Abstract
Transient receptor potential vanilloid 2 (TRPV2) was proved to play a crucial role in the tumor progression of various cancers. The association between the expression of TRPV2 and clinical outcome in cancer patients has not been studied yet. We aim to elucidate the role of TRPV2 in predicting prognosis of patients with esophageal squamous cell carcinoma (ESCC). Fresh frozen samples were collected immediately from 170 patients with ESCC after surgical resection from 2003 to 2008, including 45 pairs of tumor tissues and non-tumor tissues. TRPV2 expression was measured by quantitative real-time PCR. TRPV2 mRNA was over-expressed in ESCC tissues and cell lines. High expression of TRPV2 was observed more frequently in patients with advanced pT stage (P < 0.001), lymph node metastasis (P = 0.010) and advanced pathological stage (P = 0.001). Patients with high expression of TRPV2 (>44.40, n = 83) had worse 5-year disease-specific survival (40.0 vs 62.6 %, P < 0.001) and disease-free survival (38.4 vs 61.5 %, P < 0.001) than that with low expression (≤ 44.40, n = 87). Multivariate analysis found that the expression of TRPV2 mRNA (HR 2.19, 95 % CI 1.39-3.46, P = 0.031) and pN category (HR 2.13, 95 % CI 1.36-3.33, P = 0.001) were independent prognostic factors. Overexpression of TRPV2 mRNA was associated with poor prognosis and might serve as a novel prognostic biomarker for resected ESCC patients in early stage.
Collapse
|
111
|
Zhu Z, Liu Y, Li K, Liu J, Wang H, Sun B, Xiong Z, Jiang H, Zheng J, Hu Z. Protein tyrosine phosphatase receptor U (PTPRU) is required for glioma growth and motility. Carcinogenesis 2014; 35:1901-10. [PMID: 24876153 DOI: 10.1093/carcin/bgu123] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The membrane protein tyrosine phosphatase receptor U (PTPRU) has been shown to function as a negative regulator of adhesion and proliferation in certain cancer cell types, primarily through its dephosphorylation of β-catenin and inhibition of subsequent downstream signaling. In the present study, we set out to characterize the role of PTPRU in glioma and found that, while the expression of full-length PTPRU protein is low in these tumors, a number of non-full-length PTPRU isoforms are highly expressed. Among these isoforms, one in particular is localized to the nucleus, and its expression is increased in glioma tissues in a manner that positively correlates with malignancy grade. Short hairpin RNA knockdown of endogenous PTPRU in human and rat glioma cell lines suppressed proliferation, survival, invasion, migration, adhesion and vasculogenic tube formation in vitro, as well as intracranial tumor progression in vivo. In addition, knocking down PTPRU reduced tyrosine phosphorylation (pY) and transcriptional activity of β-catenin, and we were able to specifically rescue the cell migration defect by expressing a LEF1-β-catenin fusion protein in PTPRU-depleted cells. PTPRU knockdown also led to increased tyrosine pY of the E3 ubiquitin ligase c-Cbl and to the destabilization of several focal adhesion proteins. Taken together, our findings demonstrate that endogenous PTPRU promote glioma progression through their effect on β-catenin and focal adhesion signaling.
Collapse
Affiliation(s)
- Zhichuan Zhu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yongjie Liu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kui Li
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiwei Liu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongtao Wang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bing Sun
- Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and
| | - Zhiqi Xiong
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hualiang Jiang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Zheng
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zelan Hu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China, Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai 200030, China and Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
112
|
Zheng Q, Han L, Dong Y, Tian J, Huang W, Liu Z, Jia X, Jiang T, Zhang J, Li X, Kang C, Ren H. JAK2/STAT3 targeted therapy suppresses tumor invasion via disruption of the EGFRvIII/JAK2/STAT3 axis and associated focal adhesion in EGFRvIII-expressing glioblastoma. Neuro Oncol 2014; 16:1229-43. [PMID: 24861878 DOI: 10.1093/neuonc/nou046] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND As a commonly mutated form of the epidermal growth factor receptor, EGFRvIII strongly promotes glioblastoma (GBM) tumor invasion and progression, but the mechanisms underlying this promotion are not fully understood. METHODS Through gene manipulation, we established EGFRvIII-, wild-type EGFR-, and vector-expressing GBM cells. We used cDNA microarrays, bioinformatics analysis, target-blocking migration and invasion assays, Western blotting, and an orthotopic U87MG GBM model to examine the phenotypic shifts and treatment effects of EGFRvIII expression in vitro and in vivo. Confocal imaging, co-immunoprecipitation, and siRNA assays detected the focal adhesion-associated complex and their relationships to the EGFRvIII/JAK2/STAT3 axis in GBM cells. RESULTS The activation of JAK2/STAT3 signaling is vital for promoting migration and invasion in EGFRvIII-GBM cells. AG490 or WP1066, the JAK2/STAT3 inhibitors, specifically destroyed EGFRvIII/JAK2/STAT3-related focal adhesions and depleted the activation of EGFR/Akt/FAK and JAK2/STAT3 signaling, thereby abolishing the ability of EGFRvIII-expressing GBM cells to migrate and invade. Furthermore, the RNAi silencing of JAK2 in EGFRvIII-expressing GBM cells significantly attenuated their ability to migrate and invade; however, as a result of a potential EGFRvIII-JAK2-STAT3 activation loop, neither EGFR nor STAT3 knockdown yielded the same effects. Moreover, AG490 or JAK2 gene knockdown greatly suppressed tumor invasion and progression in the U87MG-EGFRvIII orthotopic models. CONCLUSION Taken together, our data demonstrate that JAK2/STAT3 signaling is essential for EGFRvIII-driven migration and invasion by promoting focal adhesion and stabilizing the EGFRvIII/JAK2/STAT3 axis. Targeting JAK2/STAT3 therapy, such as AG490, may have potential clinical implications for the tailored treatment of GBM patients bearing EGFRvIII-positive tumors.
Collapse
Affiliation(s)
- Qifan Zheng
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Lei Han
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Yucui Dong
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Jing Tian
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Wei Huang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Zhaoyu Liu
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Xiuzhi Jia
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Tao Jiang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Jianning Zhang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Xia Li
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Chunsheng Kang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Huan Ren
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| |
Collapse
|
113
|
Lovastatin inhibits human B lymphoma cell proliferation by reducing intracellular ROS and TRPC6 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:894-901. [PMID: 24518247 DOI: 10.1016/j.bbamcr.2014.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/17/2014] [Accepted: 02/02/2014] [Indexed: 12/21/2022]
Abstract
Clinical evidence suggests that statins reduce cancer incidence and mortality. However, there is lack of in vitro data to show the mechanism by which statins can reduce the malignancies of cancer cells. We used a human B lymphoma Daudi cells as a model and found that lovastatin inhibited, whereas exogenous cholesterol (Cho) stimulated, proliferation cell cycle progression in control Daudi cells, but not in the cells when transient receptor potential canonical 6 (TRPC6) channel was knocked down. Lovastatin decreased, whereas Cho increased, the levels of intracellular reactive oxygen species (ROS) respectively by decreasing or increasing the expression of p47-phox and gp91-phox (NOX2). Reducing intracellular ROS with either a mimetic superoxide dismutase (TEMPOL) or an NADPH oxidase inhibitor (apocynin) inhibited cell proliferation, particularly in Cho-treated cells. The effects of TEMPOL or apocynin were mimicked by inhibition of TRPC6 with SKF-96365. Lovastatin decreased TRPC6 expression and activity via a Cho-dependent mechanism, whereas Cho increased TRPC6 expression and activity via an ROS-dependent mechanism. Consistent with the fact that TRPC6 is a Ca(2+)-permeable channel, lovastatin decreased, but Cho increased, intracellular Ca(2+) also via ROS. These data suggest that lovastatin inhibits malignant B cell proliferation by reducing membrane Cho, intracellular ROS, TRPC6 expression and activity, and intracellular Ca(2+).
Collapse
|
114
|
Lei L, Lu S, Wang Y, Kim T, Mehta D, Wang Y. The role of mechanical tension on lipid raft dependent PDGF-induced TRPC6 activation. Biomaterials 2014; 35:2868-77. [PMID: 24397990 DOI: 10.1016/j.biomaterials.2013.12.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/13/2013] [Indexed: 12/22/2022]
Abstract
Canonical transient receptor potential channel 6 (TRPC6) can play an important role in governing how cells perceive the surrounding material environment and regulate Ca(2+) signaling. We have designed a TRPC6 reporter based on fluorescence resonance energy transfer (FRET) to visualize the TRPC6-mediated calcium entry and hence TRPC6 activity in live cells with high spatiotemporal resolutions. In mouse embryonic fibroblasts (MEFs), platelet-derived growth factor BB (PDGF) can activate the TRPC6 reporter, mediated by phospholipase C (PLC). This TRPC6 activation occurred mainly at lipid rafts regions of the plasma membrane because disruption of lipid raft/caveolae by methyl-β-cyclodextrin (MβCD) or the expression of dominant-negative caveolin-1 inhibited the TRPC6 activity. Culturing cells on soft materials or releasing the intracellular tension by ML-7 reduced this PDGF-induced activation of TRPC6 without affecting the PDGF-regulated Src or inositol 1,4,5-trisphosphate (IP3) receptor function, suggesting a specific role of mechanical tension in regulating TRPC6. We further showed that the release of intracellular tension had similar effect on the diffusion coefficients of TRPC6 and a raft marker, confirming a strong coupling between TRPC6 and lipid rafts. Therefore, our results suggest that the TRPC6 activation mainly occurs at lipid rafts, which is regulated by the mechanical cues of surrounding materials.
Collapse
Affiliation(s)
- Lei Lei
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States; Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Shaoying Lu
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States; Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Yi Wang
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States
| | - Taejin Kim
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States
| | - Dolly Mehta
- Department of Pharmacology, College of Medicine, University of Illinois, Chicago, IL 60612, United States
| | - Yingxiao Wang
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States; Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
115
|
Song M, Yu SP. Ionic regulation of cell volume changes and cell death after ischemic stroke. Transl Stroke Res 2013; 5:17-27. [PMID: 24323733 DOI: 10.1007/s12975-013-0314-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/16/2013] [Accepted: 11/19/2013] [Indexed: 12/20/2022]
Abstract
Stroke is a leading cause of human death and disability in the USA and around the world. Shortly after the cerebral ischemia, cell swelling is the earliest morphological change in injured neuronal, glial, and endothelial cells. Cytotoxic swelling directly results from increased Na(+) (with H2O) and Ca(2+) influx into cells via ionic mechanisms evoked by membrane depolarization and a number of harmful factors such as glutamate accumulation and the production of oxygen reactive species. During the sub-acute and chronic phases after ischemia, injured cells may show a phenotype of cell shrinkage due to complex processes involving membrane receptors/channels and programmed cell death signals. This review will introduce some progress in the understanding of the regulation of pathological cell volume changes and the involved receptors and channels, including NMDA and AMPA receptors, acid-sensing ion channels, hemichannels, transient receptor potential channels, and KCNQ channels. Moreover, accumulating evidence supports a key role of energy deficiency and dysfunction of Na(+)/K(+)-ATPase in ischemia-induced cell volume changes and cell death. Specifically, the Na(+) pump failure is a prerequisite for disruption of ionic homeostasis including a pro-apoptotic disruption of the K(+) homeostasis. Finally, we will introduce the concept of hybrid cell death as a result of the Na(+) pump failure in cultured cells and the ischemic brain. The goal of this review is to outline recent understanding of the ionic mechanism of ischemic cytotoxicity and suggest innovative ideas for future translational research.
Collapse
Affiliation(s)
- Mingke Song
- Department of Anesthesiology, Emory University School of Medicine, 101 Woodruff Circle, WMB Building Suite 620, Atlanta, GA, 30322, USA
| | | |
Collapse
|
116
|
Borowiec AS, Bidaux G, Pigat N, Goffin V, Bernichtein S, Capiod T. Calcium channels, external calcium concentration and cell proliferation. Eur J Pharmacol 2013; 739:19-25. [PMID: 24291106 DOI: 10.1016/j.ejphar.2013.10.072] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/28/2013] [Accepted: 10/17/2013] [Indexed: 11/16/2022]
Abstract
Evidence for a role for calcium channel proteins in cell proliferation is numerous suggesting that calcium influx is essential in this physiological process. Several studies in the past thirty years have demonstrated that calcium channel expression levels are determinant in cell proliferation. Voltage-gated, store-operated, second messengers and receptor-operated calcium channels have been associated to cell proliferation. However, the relationship between calcium influx and cell proliferation can be uncoupled in transformed and cancer cells, resulting in an external calcium-independent proliferation. Thus, protein expression could be more important than channel function to trigger cell proliferation suggesting that additional channel functions may be responsible to reconcile calcium channel expression and cell proliferation. When needed, external calcium concentration is obviously important for calcium channel function but it also regulates calcium sensing receptor (CaSR) activity. CaSR can up- or down-regulate cell proliferation depending on physiological conditions. CaSR sensitivity to external calcium is within the 0.5 to 5 mM range and therefore, the role of these receptors in cell proliferation must be taken into account. We therefore suggest here that cell proliferation rates could depend on the relative balance between calcium influx and CaSR activation.
Collapse
Affiliation(s)
| | - Gabriel Bidaux
- INSERM U1003, LabEx ICST, Université Lille 1, Villeneuve d'Ascq F-59655, France
| | - Natascha Pigat
- INSERM U845, Research Center Growth and Signalling Research Center, Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, Bâtiment Leriche, 96 rue Didot, Paris F-75993, France
| | - Vincent Goffin
- INSERM U845, Research Center Growth and Signalling Research Center, Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, Bâtiment Leriche, 96 rue Didot, Paris F-75993, France
| | - Sophie Bernichtein
- INSERM U845, Research Center Growth and Signalling Research Center, Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, Bâtiment Leriche, 96 rue Didot, Paris F-75993, France
| | - Thierry Capiod
- INSERM U845, Research Center Growth and Signalling Research Center, Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, Bâtiment Leriche, 96 rue Didot, Paris F-75993, France.
| |
Collapse
|
117
|
Light-controlled inhibition of malignant glioma by opsin gene transfer. Cell Death Dis 2013; 4:e893. [PMID: 24176851 PMCID: PMC3920933 DOI: 10.1038/cddis.2013.425] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 11/08/2022]
Abstract
Glioblastomas are aggressive cancers with low survival rates and poor prognosis because of their highly proliferative and invasive capacity. In the current study, we describe a new optogenetic strategy that selectively inhibits glioma cells through light-controlled membrane depolarization and cell death. Transfer of the engineered opsin ChETA (engineered Channelrhodopsin-2 variant) gene into primary human glioma cells or cell lines, but not normal astrocytes, unexpectedly decreased cell proliferation and increased mitochondria-dependent apoptosis, upon light stimulation. These optogenetic effects were mediated by membrane depolarization-induced reductions in cyclin expression and mitochondrial transmembrane potential. Importantly, the ChETA gene transfer and light illumination in mice significantly inhibited subcutaneous and intracranial glioma growth and increased the survival of the animals bearing the glioma. These results uncover an unexpected effect of opsin ion channels on glioma cells and offer the opportunity for the first time to treat glioma using a light-controllable optogenetic approach.
Collapse
|
118
|
Kim JM, Heo K, Choi J, Kim K, An W. The histone variant MacroH2A regulates Ca(2+) influx through TRPC3 and TRPC6 channels. Oncogenesis 2013; 2:e77. [PMID: 24165580 PMCID: PMC3816217 DOI: 10.1038/oncsis.2013.40] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/05/2013] [Accepted: 09/11/2013] [Indexed: 12/11/2022] Open
Abstract
The histone variant macroH2A replaces canonical H2A in the designated region of chromatin where its incorporation has the potential to establish a functionally distinct chromatin domain. The transient receptor potential canonical (TRPC) channels are a family of Ca2+-permeable cationic channels controlling changes in the cytosolic Ca2+ concentration. The proper regulation of Trpc gene expression requires chromatin remodeling, but little is known about the nature of these regulatory processes. Here, we show that macroH2A1 represses two Trpc family genes, Trpc3 and Trpc6, and attenuates Ca2+-dependent proliferative responses in bladder cancer cells. MacroH2A1 recruits histone deacetylase 1 (HDAC1) and HDAC2 to facilitate its persistent action, resulting in a compromise of histone acetylation across the Trpc3 and Trpc6 loci. Further, macroH2A1 depletion augments histone acetylation and Ca2+ influx, leading to increased cell growth and invasion. Our data provide new insights into TRPC3/TRPC6-mediated Ca2+ signaling and indicate a central role for macroH2A1 in regulating transcriptional competence of Trpc3 and Trpc6 genes.
Collapse
Affiliation(s)
- J-M Kim
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
119
|
Ay AS, Benzerdjerb N, Sevestre H, Ahidouch A, Ouadid-Ahidouch H. Orai3 constitutes a native store-operated calcium entry that regulates non small cell lung adenocarcinoma cell proliferation. PLoS One 2013; 8:e72889. [PMID: 24058448 PMCID: PMC3772818 DOI: 10.1371/journal.pone.0072889] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/21/2013] [Indexed: 11/28/2022] Open
Abstract
Orai channels have been associated with cell proliferation, survival and metastasis in several cancers. The present study investigates the expression and the role of Orai3 in cell proliferation in non-small cell lung cancer (NSCLC). We show that Orai3 is over-expressed in cancer tissues as compared to the non-tumoral ones. Furthermore, Orai3 staining is stronger in high grade tumors. Pharmacological inhibition or knockdown of Orai3 significantly reduced store operated calcium entry (SOCE), inhibited cell proliferation and arrested cells of two NSCLC cell lines in G0/G1 phase. These effects were concomitant with a down-regulation of cyclin D1, cyclin E, CDK4 and CDK2 expression. Moreover, Orai3 silencing decreased Akt phosphorylation levels. In conclusion, Orai3 constitutes a native SOCE pathway in NSCLC that controls cell proliferation and cell cycle progression likely via Akt pathway.
Collapse
Affiliation(s)
- Anne-Sophie Ay
- Laboratory of Cellular and Molecular Physiology, LPCM: EA 4667, SFR CAP-SANTE (FED 4231), UFR of Sciences, Amiens, France
| | - Nazim Benzerdjerb
- Laboratory of Cellular and Molecular Physiology, LPCM: EA 4667, SFR CAP-SANTE (FED 4231), UFR of Sciences, Amiens, France
- Pathological Anatomy and Cytology Service, Amiens North Hospital, Amiens, France
| | - Henri Sevestre
- Laboratory of Cellular and Molecular Physiology, LPCM: EA 4667, SFR CAP-SANTE (FED 4231), UFR of Sciences, Amiens, France
- Pathological Anatomy and Cytology Service, Amiens North Hospital, Amiens, France
| | - Ahmed Ahidouch
- Laboratory of Cellular and Molecular Physiology, LPCM: EA 4667, SFR CAP-SANTE (FED 4231), UFR of Sciences, Amiens, France
- Department of Biology, Ibn Zohr University, Agadir, Morocco
| | - Halima Ouadid-Ahidouch
- Laboratory of Cellular and Molecular Physiology, LPCM: EA 4667, SFR CAP-SANTE (FED 4231), UFR of Sciences, Amiens, France
- * E-mail:
| |
Collapse
|
120
|
Pdx-1 activates islet α- and β-cell proliferation via a mechanism regulated by transient receptor potential cation channels 3 and 6 and extracellular signal-regulated kinases 1 and 2. Mol Cell Biol 2013; 33:4017-29. [PMID: 23938296 DOI: 10.1128/mcb.00469-13] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The homeodomain transcription factor Pdx-1 has important roles in pancreatic development and β-cell function and survival. In the present study, we demonstrate that adenovirus-mediated overexpression of Pdx-1 in rat or human islets also stimulates cell replication. Moreover, cooverexpression of Pdx-1 with another homeodomain transcription factor, Nkx6.1, has an additive effect on proliferation compared to either factor alone, implying discrete activating mechanisms. Consistent with this, Nkx6.1 stimulates mainly β-cell proliferation, whereas Pdx-1 stimulates both α- and β-cell proliferation. Furthermore, cyclins D1/D2 are upregulated by Pdx-1 but not by Nkx6.1, and inhibition of cdk4 blocks Pdx-1-stimulated but not Nkx6.1-stimulated islet cell proliferation. Genes regulated by Pdx-1 but not Nkx6.1 were identified by microarray analysis. Two members of the transient receptor potential cation (TRPC) channel family, TRPC3 and TRPC6, are upregulated by Pdx-1 overexpression, and small interfering RNA (siRNA)-mediated knockdown of TRPC3/6 or TRPC6 alone inhibits Pdx-1-induced but not Nkx6.1-induced islet cell proliferation. Pdx-1 also stimulates extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation, an effect partially blocked by knockdown of TRPC3/6, and blockade of ERK1/2 activation with a MEK1/2 inhibitor partially impairs Pdx-1-stimulated proliferation. These studies define a pathway by which overexpression of Pdx-1 activates islet cell proliferation that is distinct from and additive to a pathway activated by Nkx6.1.
Collapse
|
121
|
Tao X, Zhao N, Jin H, Zhang Z, Liu Y, Wu J, Bast RC, Yu Y, Feng Y. FSH enhances the proliferation of ovarian cancer cells by activating transient receptor potential channel C3. Endocr Relat Cancer 2013; 20:415-29. [PMID: 23580589 PMCID: PMC3669658 DOI: 10.1530/erc-12-0005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent studies have suggested that FSH plays an important role in ovarian epithelial carcinogenesis. We demonstrated that FSH stimulates the proliferation and invasion of ovarian cancer cells, inhibits apoptosis and facilitates neovascularisation. Our previous work has shown that transient receptor potential channel C3 (TRPC3) contributes to the progression of human ovarian cancer. In this study, we further investigated the interaction between FSH and TRPC3. We found that FSH stimulation enhanced the expression of TRPC3 at both the mRNA and protein levels. siRNA-mediated silencing of TRPC3 expression inhibited the ability of FSH to stimulate proliferation and blocked apoptosis in ovarian cancer cell lines. FSH stimulation was associated with the up-regulation of TRPC3, while also facilitating the influx of Ca(2)(+) after treatment with a TRPC-specific agonist. Knockdown of TRPC3 abrogated FSH-stimulated Akt/PKB phosphorylation, leading to decreased expression of downstream effectors including survivin, HIF1-α and VEGF. Ovarian cancer specimens were analysed for TRPC3 expression; higher TRPC3 expression levels correlated with early relapse and worse prognosis. Association with poor disease-free survival and overall survival remained after adjusting for clinical stage and grade. In conclusion, TRPC3 plays a significant role in the stimulating activity of FSH and could be a potential therapeutic target for the treatment of ovarian cancer, particularly in postmenopausal women with elevated FSH levels.
Collapse
Affiliation(s)
- Xiang Tao
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Naiqing Zhao
- Department of Biostatistics, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hongyan Jin
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Zhenbo Zhang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital of Jiao Tong University, Shanghai 200080, China
| | - Yintao Liu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Jian Wu
- Department of Pathology, Gongli Hospital, Shanghai, 200135, China
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Yinhua Yu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- To whom correspondence and offprint requests should be addressed: Youji Feng, Department of Gynecology, Obstetrics and Gynecology, Hospital of Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology, Shanghai First People's Hospital of Jiao Tong University, Shanghai 200080, China. Phone: 8621-63240090-3082; Fax: 8621-63241377; Yinhua Yu, Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China. Phone: 8621-33189900; Fax: 8621-63455090;
| | - Youji Feng
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital of Jiao Tong University, Shanghai 200080, China
- To whom correspondence and offprint requests should be addressed: Youji Feng, Department of Gynecology, Obstetrics and Gynecology, Hospital of Fudan University, Shanghai 200011, China; Department of Obstetrics and Gynecology, Shanghai First People's Hospital of Jiao Tong University, Shanghai 200080, China. Phone: 8621-63240090-3082; Fax: 8621-63241377; Yinhua Yu, Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China. Phone: 8621-33189900; Fax: 8621-63455090;
| |
Collapse
|
122
|
Song J, Wang Y, Li X, Shen Y, Yin M, Guo Y, Diao L, Liu Y, Yue D. Critical role of TRPC6 channels in the development of human renal cell carcinoma. Mol Biol Rep 2013; 40:5115-22. [PMID: 23700295 DOI: 10.1007/s11033-013-2613-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Accepted: 04/30/2013] [Indexed: 02/02/2023]
Abstract
Renal cell carcinoma (RCC) is the most common tumor arising from the cells in the lining of tubules in the kidney. Some members of the Ca2+-permeable transient receptor potential canonical (TRPC) family of channel proteins have demonstrated a role in the proliferation of some types of cancer cells. In this study, we investigated the role of TRPC6 in the development of human RCC. RT-PCR and Western blotting were used to investigate TRPC6 expression in 1932 and ACHN cells. Immunohistochemical techniques were applied to study TRPC6 expression in 60 cases of RCC primary tissue samples and 10 cases of corresponding normal renal tissues. To inhibit TRPC6 activity or expression, RNA interference was used. The effects of TRPC6 channels on RCC cell viability and cell cycle progression were investigated by MTT and flow cytometry. TRPC6 was expressed in 1932 and ACHN cells. TRPC6 protein was detected in 73.3% of RCC samples, and there was a significant difference compared with the normal renal samples (30%) (p<0.05). Moreover the level of TRPC6 expression was associated with RCC Fuhrman grade (p<0.01). Blockade of TRPC6 channels in ACHN cells suppressed basal cell proliferation and partially inhibited HGF-induced cell proliferation. Furthermore, inhibition of TRPC6 channels expression prolonged the transition through G2/M phase in ACHN cells. In summary, expression of TRPC6 is markedly increased in RCC specimens and associated with RCC histological grade. TRPC6 plays an important role in ACHN cells proliferation.
Collapse
Affiliation(s)
- Jiayin Song
- School of Laboratory Medicine, Tianjin Medical University, Tianjin, 300203, China
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
High expression of transient potential receptor C6 correlated with poor prognosis in patients with esophageal squamous cell carcinoma. Med Oncol 2013; 30:607. [PMID: 23686700 DOI: 10.1007/s12032-013-0607-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 05/10/2013] [Indexed: 02/07/2023]
Abstract
TRPC6 plays a crucial role in the tumor progression of various cancers. The relation between the expression of TRPC6 and clinical prognosis has not been studied yet. Our study was to elucidate the role of TRPC6 in predicting outcomes of patients with esophageal squamous cell carcinoma (ESCC). Fresh frozen samples were collected immediately from 172 patients with ESCC after surgical resection from 2003 to 2008 at Sun Yat-sen University Cancer Center, including 45 pairs of tumor tissues and nontumor tissues. TRPC6 expression was measured by quantitative real-time PCR and Western blotting analyses. TRPC6 mRNA and protein were up-regulated in ESCC tissues when compared with the paired nontumor tissues. High expression of TRPC6 mRNA was associated with the higher pT status (P = 0.016) and pathological staging (P = 0.040). The 5-year disease-specific survival in the high expression of TRPC6 mRNA group (>188.98, n = 81) is poorer than that in low-level expression group (≤188.98, n = 91) (42.1 vs. 62.7 %, P = 0.004). Stratified analysis according to the pathological stage revealed its discernibility on DSS was only pronounced in patients with pStage III (P = 0.015). Cox multivariate analysis revealed that pN category (P < 0.001; Relative risk, 2.897, 95 % CI 1.830-4.585) and the expression of TRPC6 mRNA (P = 0.006; Relative risk, 1.863, 95 % CI 1.196-2.902) were independent prognostic factors. TRPC6 mRNA overexpression correlated with poor prognosis in patients with ESCC and might serve as a novel prognostic biomarker for resected ESCC patients in advanced stage.
Collapse
|
124
|
Chen YF, Chen YT, Chiu WT, Shen MR. Remodeling of calcium signaling in tumor progression. J Biomed Sci 2013; 20:23. [PMID: 23594099 PMCID: PMC3639169 DOI: 10.1186/1423-0127-20-23] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 04/08/2013] [Indexed: 02/06/2023] Open
Abstract
Intracellular Ca2+ is one of the crucial signalings that modulate various cellular functions. The dysregulation of Ca2+ homeostasis has been suggested as an important event in driving the expression of the malignant phenotypes, such as proliferation, migration, invasion, and metastasis. Cell migration is an early prerequisite for tumor metastasis that has a significant impact on patient prognosis. During cell migration, the exquisite spatial and temporal organization of intracellular Ca2+ provides a rapid and robust way for the selective activation of signaling components that play a central role in cytoskeletal reorganization, traction force generation, and focal adhesion dynamics. A number of known molecular components involved in Ca2+ influx pathways, including stromal interaction molecule (STIM)/Orai-mediated store-operated Ca2+ entry (SOCE) and the Ca2+-permeable transient receptor potential (TRP) channels, have been implicated in cancer cell migration and tumor metastasis. The clinical significance of these molecules, such as STIM proteins and the TRPM7 channel, in tumor progression and their diagnostic and prognostic potentials have also been demonstrated in specific cancer types. In this review, we summarize the recent advances in understanding the important roles and regulatory mechanisms of these Ca2+ influx pathways on malignant behaviors of tumor cells. The clinical implications in facilitating current diagnostic and therapeutic procedures are also discussed.
Collapse
Affiliation(s)
- Yih-Fung Chen
- Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | | | | | | |
Collapse
|
125
|
Barrantes-Freer A, Kim E, Bielanska J, Giese A, Mortensen LS, Schulz-Schaeffer WJ, Stadelmann C, Brück W, Pardo LA. Human glioma-initiating cells show a distinct immature phenotype resembling but not identical to NG2 glia. J Neuropathol Exp Neurol 2013; 72:307-24. [PMID: 23481707 PMCID: PMC3678885 DOI: 10.1097/nen.0b013e31828afdbd] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Glioma-initiating cells (GICs) represent a potential important therapeutic target because they are likely to account for the frequent recurrence of malignant gliomas; however, their identity remains unsolved. Here, we characterized the cellular lineage fingerprint of GICs through a combination of electrophysiology, lineage marker expression, and differentiation assays of 5 human patient-derived primary GIC lines. Most GICs coexpressed nestin, NG2 proteoglycan, platelet-derived growth factor receptor-α, and glial fibrillary acidic protein. Glioma-initiating cells could be partially differentiated into astrocytic but not oligodendroglial or neural lineages. We also demonstrate that GICs have a characteristic electrophysiologic profile distinct from that of well-characterized tumor bulk cells. Together, our results suggest that GICs represent a unique type of cells reminiscent of an immature phenotype that closely resembles but is not identical to NG2 glia with respect to marker expression and functional membrane properties.
Collapse
Affiliation(s)
- Alonso Barrantes-Freer
- Max-Planck-Institute of Experimental Medicine, Molecular Biology of Neuronal Signals, AG Oncophysiology, Göttingen
| | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Bernaldo de Quirós S, Merlo A, Secades P, Zambrano I, de Santa María IS, Ugidos N, Jantus-Lewintre E, Sirera R, Suarez C, Chiara MD. Identification of TRPC6 as a possible candidate target gene within an amplicon at 11q21-q22.2 for migratory capacity in head and neck squamous cell carcinomas. BMC Cancer 2013; 13:116. [PMID: 23497198 PMCID: PMC3606258 DOI: 10.1186/1471-2407-13-116] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 03/07/2013] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cytogenetic and gene expression analyses in head and neck squamous cell carcinomas (HNSCC) have allowed identification of genomic aberrations that may contribute to cancer pathophysiology. Nevertheless, the molecular consequences of numerous genetic alterations still remain unclear. METHODS To identify novel genes implicated in HNSCC pathogenesis, we analyzed the genomic alterations present in five HNSCC-derived cell lines by array CGH, and compared high level focal gene amplifications with gene expression levels to identify genes whose expression is directly impacted by these genetic events. Next, we knocked down TRPC6, one of the most highly amplified and over-expressed genes, to characterize the biological roles of TRPC6 in carcinogenesis. Finally, real time PCR was performed to determine TRPC6 gene dosage and mRNA levels in normal mucosa and human HNSCC tissues. RESULTS The data showed that the HNSCC-derived cell lines carry most of the recurrent genomic abnormalities previously described in primary tumors. High-level genomic amplifications were found at four chromosomal sites (11q21-q22.2, 18p11.31-p11.21, 19p13.2-p13.13, and 21q11) with associated gene expression changes in selective candidate genes suggesting that they may play an important role in the malignant behavior of HNSCC. One of the most dramatic alterations of gene transcription involved the TRPC6 gene (located at 11q21-q22.2) which has been recently implicated in tumour invasiveness. siRNA-induced knockdown of TRPC6 expression in HNSCC-derived cells dramatically inhibited HNSCC-cell invasion but did not significantly alter cell proliferation. Importantly, amplification and concomitant overexpression of TRPC6 was also found in HNSCC tumour samples. CONCLUSIONS Altogether, these data show that TRPC6 is likely to be a target for 11q21-22.2 amplification that confers enhanced invasive behavior to HNSCC cells. Therefore, TRPC6 may be a promising therapeutic target in the treatment of HNSCC.
Collapse
Affiliation(s)
- Sandra Bernaldo de Quirós
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Kim SY, Yang D, Myeong J, Ha K, Kim SH, Park EJ, Kim IG, Cho NH, Lee KP, Jeon JH, So I. Regulation of calcium influx and signaling pathway in cancer cells via TRPV6-Numb1 interaction. Cell Calcium 2013; 53:102-11. [PMID: 23140583 DOI: 10.1016/j.ceca.2012.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 10/10/2012] [Accepted: 10/12/2012] [Indexed: 11/19/2022]
Abstract
Ca(2+) is a critical factor in the regulation of signal transduction and Ca(2+) homeostasis is altered in different human diseases. The level of Ca(2+) in cells is highly regulated through a diverse class of regulators. Among them is the transient receptor potential vanilloid 6 (TRPV6), which is a Ca(2+) selective channel that absorbs Ca(2+) in the small intestine. TRPV6 is overexpressed in some cancers and exhibits oncogenic potential, but its exact mechanism is still poorly understood. The Numb protein is a cell fate determinant that functions in endocytosis and as a tumor suppressor via the stabilization of p53. Numb protein consisted of four isoforms. Here, we showed a novel function of Numb1, which negatively regulates TRPV6 activity. The expression of Numb1 decreased cytosolic Ca(2+) concentrations in TRPV6-transfected HEK293 cells. When all the isoforms of Numb were depleted using siRNA in a TRPV6 stable cell line, the levels of cytosolic Ca(2+) increased. We observed an interaction between Numb1 and TRPV6 using co-immunoprecipitation. We confirmed this interaction using Fluorescence Resolution Energy Transfer (FRET). We identified the TRPV6 and Numb1 binding site using TRPV6 C-terminal truncation mutants and Numb1 deletion mutants. The binding site in TRPV6 was an aspartic acid at amino acid residue 716, and that binding site in Numb1 was arginine at amino acid residue 434. A Numb1 mutant, lacking TRPV6 binding activity, failed to inhibit TRPV6 activity. Every isoform of Numb knockdown, using an siRNA-based approach in MCF-7 breast cancer cells, not only showed enhanced TRPV6 expression but also both the cytosolic Ca(2+) concentration and cell proliferation were increased. The down-regulated expression of TRPV6 using siRNA increased Numb protein expression; however, the cytosolic influx of Ca(2+) and proliferation of the cell were decreased. To examine downstream signaling during Ca(2+) influx, we performed Western blotting analysis on TRPV6 upregulated cancer cells (MCF-7, PC-3). Taken together, these results demonstrated that Numb1 interacts with TRPV6 through charged residues and inhibits its activity via the regulation of protein expression. Moreover, we provided evidence for a Ca(2+)-regulated cancer cell signaling pathway and that the Ca(2+) channel is a target of cancer cells.
Collapse
Affiliation(s)
- Sung-Young Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
TRP channels: diagnostic markers and therapeutic targets for breast cancer? Trends Mol Med 2013; 19:117-24. [DOI: 10.1016/j.molmed.2012.11.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 09/25/2012] [Accepted: 11/13/2012] [Indexed: 01/22/2023]
|
129
|
TRPC5 channel is the mediator of neurotrophin-3 in regulating dendritic growth via CaMKIIα in rat hippocampal neurons. J Neurosci 2012; 32:9383-95. [PMID: 22764246 DOI: 10.1523/jneurosci.6363-11.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neurotrophin-3 (NT-3) plays numerous important roles in the CNS and the elevation of intracellular Ca(2+) ([Ca(2+)](i)) is critical for these functions of NT-3. However, the mechanism by which NT-3 induces [Ca(2+)](i) elevation remains largely unknown. Here, we found that transient receptor potential canonical (TRPC) 5 protein and TrkC, the NT-3 receptor, exhibited a similar temporal expression in rat hippocampus and cellular colocalization in hippocampal neurons. Stimulation of the neurons by NT-3 induced a nonselective cation conductance and PLCγ-dependent [Ca(2+)](i) elevation, which were both blocked when TRPC5, but not TRPC6 channels, were inhibited. Moreover, the Ca(2+) influx through TRPC5 induced by NT-3 inhibited the neuronal dendritic growth through activation of calmodulin-dependent kinase (CaMK) IIα. In contrast, the Ca(2+) influx through TRPC6 induced by NT-4 promoted the dendritic growth. Thus, TRPC5 acts as a novel and specific mediator for NT-3 to regulate dendrite development through CaMKIIα.
Collapse
|
130
|
[TRP calcium channel and breast cancer: expression, role and correlation with clinical parameters]. Bull Cancer 2012; 99:655-64. [PMID: 22640890 DOI: 10.1684/bdc.2012.1595] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Breast cancer (BC) has the highest incidence rate in women in industrialized countries. Statistically, it is estimated that one out of 10 women will develop BC during her life. Evidence is accumulating for the role of ion channels in the development of cancer. Most studied ion channels in BC are K(+) channels, which are involved in cell proliferation, cell cycle progression and cell migration, and Na(+) channels, which correlate with invasiveness. Emerging studies demonstrated the role of Ca(2+) signaling in cancer cell proliferation, survival and migration. Recent findings demonstrated that the expression and/or activity of the transient receptor potential (TRP) channels are altered in several cancers. Among the TRP families, TRPC (canonical or classical), TRPM (melastatin) and TRPV (vanilloid) are related to malignant growth and cancer progression. Although these channels are frequently and abundantly expressed in many tumors, their specific expression, activity and roles in BC are still poorly understood. The expression of TRP channels has also been proposed as a tool for diagnosis, prognosis and/or therapeutic issues of several diseases. In cancer, TRPV6 and TRPM8 have been proposed as tumor progression markers of prostate cancer outcome and TRPC6 as a novel therapeutic target for esophageal carcinoma. Interestingly high levels of TRPC3 expression correlate with a favorable prognosis in patients with lung adenocarcinoma. Our team has recently reported the expression and role of TRPC1, TRPC6, TRPM7, TRPM8 and TRPV6 in BC cell lines and primary cultures. We have also investigated TRP expression and their clinical significance in human breast adenocarcinoma and we suggest that TRP channels are new potential BC markers. Indeed TRPC1 and TRPM8 may be considered as good prognosis markers of well-differentiated tumors, TRPM7 as a proliferative marker of poorly differentiated tumors and TRPV6 as a prognosis marker of aggressive cancers. In this review, we summarize the data reported to date regarding the changes in TRP expression associated with BC. We also discuss the importance of TRP channels in BC cells proliferation and migration and their interest as new BC markers.
Collapse
|
131
|
Zhao J, He H, Zhou K, Ren Y, Shi Z, Wu Z, Wang Y, Lu Y, Jiao J. Neuronal transcription factors induce conversion of human glioma cells to neurons and inhibit tumorigenesis. PLoS One 2012; 7:e41506. [PMID: 22859994 PMCID: PMC3409237 DOI: 10.1371/journal.pone.0041506] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 06/22/2012] [Indexed: 01/09/2023] Open
Abstract
Recent findings have demonstrated that the overexpression of lineage-specific transcription factors induces cell fate changes among diverse cell types. For example, neurons can be generated from mouse and human fibroblasts. It is well known that neurons are terminally differentiated cells that do not divide. Therefore, we consider how to induce glioma cells to become neurons by introducing transcription factors. Here, we describe the efficient generation of induced neuronal (iN) cells from glioma cells by the infection with three transcription factors: Ascl1, Brn2 and Ngn2 (ABN). iN cells expressed multiple neuronal markers and fired action potentials, similar to the properties of authentic neurons. Importantly, the proliferation of glioma cells following ABN overexpression was dramatically inhibited in both in vitro and in vivo experiments. In addition, iN cells that originated from human glioma cells did not continue to grow when they were sorted and cultured in vitro. The strategies by which glioma cells are induced to become neurons may be used to clinically study methods for inhibiting tumor growth.
Collapse
Affiliation(s)
- Junli Zhao
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Hua He
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Kechun Zhou
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yonggang Ren
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Zixiao Shi
- Institute of Zoology, State Key Laboratory of Reproductive Biology, Chinese Academy of Sciences, Beijing, China
| | | | - Yizheng Wang
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yicheng Lu
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
- * E-mail: (YL); (JJ)
| | - Jianwei Jiao
- Institute of Zoology, State Key Laboratory of Reproductive Biology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (YL); (JJ)
| |
Collapse
|
132
|
Monteith GR, Davis FM, Roberts-Thomson SJ. Calcium channels and pumps in cancer: changes and consequences. J Biol Chem 2012; 287:31666-73. [PMID: 22822055 DOI: 10.1074/jbc.r112.343061] [Citation(s) in RCA: 295] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Increases in intracellular free Ca(2+) play a major role in many cellular processes. The deregulation of Ca(2+) signaling is a feature of a variety of diseases, and modulators of Ca(2+) signaling are used to treat conditions as diverse as hypertension to pain. The Ca(2+) signal also plays a role in processes important in cancer, such as proliferation and migration. Many studies in cancer have identified alterations in the expression of proteins involved in the movement of Ca(2+) across the plasma membrane and subcellular organelles. In some cases, these Ca(2+) channels or pumps are potential therapeutic targets for specific cancer subtypes or correlate with prognosis.
Collapse
Affiliation(s)
- Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane 4075, Australia.
| | | | | |
Collapse
|
133
|
Expression and Role of the Intermediate-Conductance Calcium-Activated Potassium Channel KCa3.1 in Glioblastoma. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:421564. [PMID: 22675627 PMCID: PMC3362965 DOI: 10.1155/2012/421564] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 03/15/2012] [Indexed: 12/29/2022]
Abstract
Glioblastomas are characterized by altered expression of several ion channels that have important consequences in cell functions associated with their aggressiveness, such as cell survival, proliferation, and migration. Data on the altered expression and function of the intermediate-conductance calcium-activated K (KCa3.1) channels in glioblastoma cells have only recently become available. This paper aims to (i) illustrate the main structural, biophysical, pharmacological, and modulatory properties of the KCa3.1 channel, (ii) provide a detailed account of data on the expression of this channel in glioblastoma cells, as compared to normal brain tissue, and (iii) critically discuss its major functional roles. Available data suggest that KCa3.1 channels (i) are highly expressed in glioblastoma cells but only scantly in the normal brain parenchima, (ii) play an important role in the control of glioblastoma cell migration. Altogether, these data suggest KCa3.1 channels as potential candidates for a targeted therapy against this tumor.
Collapse
|
134
|
Shankaran H, Weber TJ, von Neubeck C, Sowa MB. Using imaging methods to interrogate radiation-induced cell signaling. Radiat Res 2012; 177:496-507. [PMID: 22380462 DOI: 10.1667/rr2669.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
There is increasing emphasis on the use of systems biology approaches to define radiation-induced responses in cells and tissues. Such approaches frequently rely on global screening using various high throughput 'omics' platforms. Although these methods are ideal for obtaining an unbiased overview of cellular responses, they often cannot reflect the inherent heterogeneity of the system or provide detailed spatial information. Additionally, performing such studies with multiple sampling time points can be prohibitively expensive. Imaging provides a complementary method with high spatial and temporal resolution capable of following the dynamics of signaling processes. In this review, we utilize specific examples to illustrate how imaging approaches have furthered our understanding of radiation-induced cellular signaling. Particular emphasis is placed on protein colocalization, and oscillatory and transient signaling dynamics.
Collapse
Affiliation(s)
- Harish Shankaran
- Computational Biology and Bioinformatics, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | | | | | | |
Collapse
|
135
|
Abstract
Ion channels and G-protein-coupled receptors (GPCRs) play a fundamental role in cancer progression by influencing Ca(2+) influx and signaling pathways in transformed cells. Transformed cells thrive in a hostile environment that is characterized by extracellular acidosis that promotes the pathological phenotype. The pathway(s) by which extracellular protons achieve this remain unclear. Here, a role for proton-sensing ion channels and GPCRs as mediators of the effects of extracellular protons in cancer cells is discussed.
Collapse
Affiliation(s)
- Maike Glitsch
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom.
| |
Collapse
|
136
|
Molenaar RJ. Ion channels in glioblastoma. ISRN NEUROLOGY 2011; 2011:590249. [PMID: 22389824 PMCID: PMC3263536 DOI: 10.5402/2011/590249] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 09/19/2011] [Indexed: 11/23/2022]
Abstract
Glioblastoma is the most common primary brain tumor with the most dismal prognosis. It is characterized by extensive invasion, migration, and angiogenesis. Median survival is only 15 months due to this behavior, rendering focal surgical resection ineffective and adequate radiotherapy impossible. At this moment, several ion channels have been implicated in glioblastoma proliferation, migration, and invasion. This paper summarizes studies on potassium, sodium, chloride, and calcium channels of glioblastoma. It provides an up-to-date overview of the literature that could ultimately lead to new therapeutic targets.
Collapse
Affiliation(s)
- Remco J Molenaar
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
137
|
Effect of thymol on Ca2+ homeostasis and viability in human glioblastoma cells. Eur J Pharmacol 2011; 670:85-91. [PMID: 21914442 DOI: 10.1016/j.ejphar.2011.08.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 07/27/2011] [Accepted: 08/17/2011] [Indexed: 01/29/2023]
Abstract
The effect of the natural essential oil thymol on cytosolic Ca(2+) concentrations ([Ca(2+)](i)) and viability in human glioblastoma cells was examined. The Ca(2+)-sensitive fluorescent dye fura-2 was applied to measure [Ca(2+)](i). Thymol at concentrations of 400-1000 μM induced a [Ca(2+)](i) rise in a concentration-dependent fashion. The response was decreased partially by removal of extracellular Ca(2+). Thymol-induced Ca(2+) signal was not altered by nifedipine, econazole, SK&F96365, and protein kinase C activator phorbol myristate acetate (PMA), but was inhibited by the protein kinase C inhibitor GF109203X. When extracellular Ca(2+) was removed, incubation with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) abolished thymol-induced [Ca(2+)](i) rise. Incubation with thymol also abolished thapsigargin or BHQ-induced [Ca(2+)](i) rise. Inhibition of phospholipase C with U73122 abolished thymol-induced [Ca(2+)](i) rise. At concentrations of 200-800 μM, thymol killed cells in a concentration-dependent manner. This cytotoxic effect was not changed by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/acetoxy methyl (BAPTA/AM). Annexin V/propidium iodide staining data suggest that thymol (200, 400 and 600 μM) induced apoptosis in a concentration-dependent manner. Collectively, in human glioblastoma cells, thymol induced a [Ca(2+)](i) rise by inducing phospholipase C- and protein kinase C-dependent Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry via non store-operated Ca(2+) channels. Thymol induced cell death that may involve apoptosis.
Collapse
|
138
|
Becchetti A. Ion channels and transporters in cancer. 1. Ion channels and cell proliferation in cancer. Am J Physiol Cell Physiol 2011; 301:C255-65. [DOI: 10.1152/ajpcell.00047.2011] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Progress through the cell mitotic cycle requires precise timing of the intrinsic molecular steps and tight coordination with the environmental signals that maintain a cell into the proper physiological context. Because of their great functional flexibility, ion channels coordinate the upstream and downstream signals that converge on the cell cycle machinery. Both voltage- and ligand-gated channels have been implicated in the control of different cell cycle checkpoints in normal as well as neoplastic cells. Ion channels mediate the calcium signals that punctuate the mitotic process, the cell volume oscillations typical of cycling cells, and the exocytosis of autocrine or angiogenetic factors. Other functions of ion channels in proliferation are still matter of debate. These may or may not depend on ion transport, as the channel proteins can form macromolecular complexes with growth factor and cell adhesion receptors. Direct conformational coupling with the cytoplasmic regulatory proteins is also possible. Derangement or relaxed control of the above processes can promote neoplasia. Specific types of ion channels have turned out to participate in the different stages of the tumor progression, in which cell heterogeneity is increased by the selection of malignant cell clones expressing the ion channel types that better support unrestrained growth. However, a comprehensive mechanistic picture of the functional relations between ion channels and cell proliferation is yet not available, partly because of the considerable experimental challenges offered by studying these processes in living mammalian cells. No doubt, such studies will constitute one of the most fruitful research fields for the next generation of cell physiologists.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
139
|
Ritchie MF, Zhou Y, Soboloff J. WT1/EGR1-mediated control of STIM1 expression and function in cancer cells. Front Biosci (Landmark Ed) 2011; 16:2402-15. [PMID: 21622185 DOI: 10.2741/3862] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There have been numerous publications linking Ca(2+) signaling and cancer, however, a clear explanation for this link has remained elusive. We recently identified the oncogenes/tumor suppressors Wilms Tumor Suppressor 1 (WT1) and Early Growth Response 1 (EGR1) as regulators of the expression of STIM1, an essential regulator of Ca(2+) entry in non-excitable cells. The current review focuses on the literature defining both differential Ca(2+) signaling and WT1/EGR1 expression patterns in 6 specific cancer subtypes: Acute Myeloid Leukemia, Wilms Tumor, breast cancer, ovarian cancer, glioblastoma and prostate cancer. For each tumor-type, we have assessed how specific changes in WT1 and EGR1 expression might contribute to aberrant Ca(2+) homeostasis as well as the therapeutic potential of these observations.
Collapse
Affiliation(s)
- Michael F Ritchie
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | |
Collapse
|
140
|
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: 3.4] [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
|
141
|
Ambatipudi S, Gerstung M, Gowda R, Pai P, Borges AM, Schäffer AA, Beerenwinkel N, Mahimkar MB. Genomic profiling of advanced-stage oral cancers reveals chromosome 11q alterations as markers of poor clinical outcome. PLoS One 2011; 6:e17250. [PMID: 21386901 PMCID: PMC3046132 DOI: 10.1371/journal.pone.0017250] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 01/22/2011] [Indexed: 11/19/2022] Open
Abstract
Identifying oral cancer lesions associated with high risk of relapse and predicting clinical outcome remain challenging questions in clinical practice. Genomic alterations may add prognostic information and indicate biological aggressiveness thereby emphasizing the need for genome-wide profiling of oral cancers. High-resolution array comparative genomic hybridization was performed to delineate the genomic alterations in clinically annotated primary gingivo-buccal complex and tongue cancers (n = 60). The specific genomic alterations so identified were evaluated for their potential clinical relevance. Copy-number changes were observed on chromosomal arms with most frequent gains on 3q (60%), 5p (50%), 7p (50%), 8q (73%), 11q13 (47%), 14q11.2 (47%), and 19p13.3 (58%) and losses on 3p14.2 (55%) and 8p (83%). Univariate statistical analysis with correction for multiple testing revealed chromosomal gain of region 11q22.1–q22.2 and losses of 17p13.3 and 11q23–q25 to be associated with loco-regional recurrence (P = 0.004, P = 0.003, and P = 0.0003) and shorter survival (P = 0.009, P = 0.003, and P 0.0001) respectively. The gain of 11q22 and loss of 11q23-q25 were validated by interphase fluorescent in situ hybridization (I-FISH). This study identifies a tractable number of genomic alterations with few underlying genes that may potentially be utilized as biological markers for prognosis and treatment decisions in oral cancers.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cells, Cultured
- Chromosome Aberrations
- Chromosomes, Human, Pair 11/genetics
- Comparative Genomic Hybridization
- Disease Progression
- Female
- Gene Expression Profiling
- Genome, Human
- HeLa Cells
- Humans
- In Situ Hybridization, Fluorescence
- Male
- Middle Aged
- Mouth Neoplasms/diagnosis
- Mouth Neoplasms/genetics
- Mouth Neoplasms/pathology
- Neoplasm Staging
- Prognosis
- Validation Studies as Topic
Collapse
Affiliation(s)
- Srikant Ambatipudi
- Tata Memorial Centre (TMC), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Cancer Research Institute (CRI), Navi Mumbai, India
| | - Moritz Gerstung
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Ravindra Gowda
- Tata Memorial Centre (TMC), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Cancer Research Institute (CRI), Navi Mumbai, India
| | - Prathamesh Pai
- Head and Neck Unit, Tata Memorial Hospital, Tata Memorial Centre (TMC), Mumbai, India
| | - Anita M. Borges
- Department of Pathology and Laboratory Medicine, S. L. Raheja Hospital, Mumbai, India
| | - Alejandro A. Schäffer
- Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, Maryland, United States of America
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Manoj B. Mahimkar
- Tata Memorial Centre (TMC), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Cancer Research Institute (CRI), Navi Mumbai, India
- * E-mail:
| |
Collapse
|
142
|
Dhennin-Duthille I, Gautier M, Faouzi M, Guilbert A, Brevet M, Vaudry D, Ahidouch A, Sevestre H, Ouadid-Ahidouch H. High Expression of Transient Receptor Potential Channels in Human Breast Cancer Epithelial Cells and Tissues: Correlation with Pathological Parameters. Cell Physiol Biochem 2011; 28:813-22. [DOI: 10.1159/000335795] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2011] [Indexed: 01/19/2023] Open
|
143
|
Parkash J, Asotra K. Calcium wave signaling in cancer cells. Life Sci 2010; 87:587-95. [PMID: 20875431 DOI: 10.1016/j.lfs.2010.09.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 09/16/2010] [Indexed: 02/05/2023]
Abstract
Ca(2+) functions as an important signaling messenger right from beginning of life to the final moments of the end of life. Ca(2+) is needed at several steps of the cell cycle such as early G(1), at the G(1)/S, and G(2)/M transitions. The Ca(2+) signals in the form of time-dependent changes in intracellular Ca(2+) concentrations, [Ca(2+)](i), are presented as brief spikes organized into regenerative Ca(2+) waves. Ca(2+)-mediated signaling pathways have also been shown to play important roles in carcinogenesis such as transformation of normal cells to cancerous cells, tumor formation and growth, invasion, angiogenesis and metastasis. Since the global Ca(2+) oscillations arise from Ca(2+) waves initiated locally, it results in stochastic oscillations because although each cell has many IP(3)Rs and Ca(2+) ions, the law of large numbers does not apply to the initiating event which is restricted to very few IP(3)Rs due to steep Ca(2+) concentration gradients. The specific Ca(2+) signaling information is likely to be encoded in a calcium code as the amplitude, duration, frequency, waveform or timing of Ca(2+) oscillations and decoded again at a later stage. Since Ca(2+) channels or pumps involved in regulating Ca(2+) signaling pathways show altered expression in cancer, one can target these Ca(2+) channels and pumps as therapeutic options to decrease proliferation of cancer cells and to promote their apoptosis. These studies can provide novel insights into alterations in Ca(2+) wave patterns in carcinogenesis and lead to the development of newer technologies based on Ca(2+) waves for the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Jai Parkash
- Robert Stempel College of Public Health and Social Work, Department of Environmental and Occupational Health, Florida International University, 11200 SW 8th Street, HLS-594, Miami, FL 33199, USA.
| | | |
Collapse
|