51
|
Sharifi N, Salmaninejad A, Ferdosi S, Bajestani AN, Khaleghiyan M, Estiar MA, Jamali M, Nowroozi MR, Shakoori A. HER2 gene amplification in patients with prostate cancer: Evaluating a CISH-based method. Oncol Lett 2016; 12:4651-4658. [PMID: 28105172 DOI: 10.3892/ol.2016.5235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 07/12/2016] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer (PCa) is one of the most widespread malignancies in the world. The role of the human epidermal growth factor receptor 2 (HER2) in the pathogenesis and progression of human PCa remains poorly understood. In contradiction with breast cancer, studies on HER2 overexpression and gene amplification in PCa have produced varying results, although the HER2 oncogene has been implicated in the biology of numerous tumor types, and serves as a prognostic marker and therapeutic target in breast cancer. Technical challenges are considered the main reasons for data discrepancies. Amplification of the HER2 gene has previously been reported in PCa, in which it was associated with tumor progression. The present study aimed to evaluate the prevalence and clinical significance of HER2 amplification in PCa. A total of 32 biopsy samples obtained from human prostate adenocarcinomas were evaluated by chromogenic in situ hybridization (CISH) to determine the frequency of patients with HER2 gene amplifications. High copy numbers of HER2 were detected in 19 of the prostate tumors analyzed. The results of the present study suggested that, in patients without amplification of HER2, high levels of prostate-specific antigen or a high Gleason score were not significantly correlated with a high pathologic stage. Furthermore, amplification levels of the HER2 gene were directly associated with pathologic stage in patients with PCa. Therefore, the potential use of HER2 as a prognostic factor or therapeutic target for PCa warrants further study.
Collapse
Affiliation(s)
- Nazanin Sharifi
- Department of Medical Genetics, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Arash Salmaninejad
- Department of Medical Genetics, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran; Genetic Research Center, Student Research Committee, Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Samira Ferdosi
- Department of Medical Genetics, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Abolfazl Nesaei Bajestani
- Department of Medical Genetics, Ayatollah Madani Hospital, Gonabad University of Medical Sciences, Gonabad 9698154813, Iran
| | - Malihe Khaleghiyan
- Department of Medical Genetics, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran; Department of Medical Genetics, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Mehrdad Asghari Estiar
- Department of Medical Genetics, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Mansour Jamali
- Department of Pathology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Mohammad Reza Nowroozi
- Uro Oncology Research Center, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| | - Abbas Shakoori
- Department of Medical Genetics, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran; Department of Medical Genetics, Tehran University of Medical Sciences, Tehran 1471613151, Iran
| |
Collapse
|
52
|
Cheng YY, Wright CM, Kirschner MB, Williams M, Sarun KH, Sytnyk V, Leshchynska I, Edelman JJ, Vallely MP, McCaughan BC, Klebe S, van Zandwijk N, Lin RCY, Reid G. KCa1.1, a calcium-activated potassium channel subunit alpha 1, is targeted by miR-17-5p and modulates cell migration in malignant pleural mesothelioma. Mol Cancer 2016; 15:44. [PMID: 27245839 PMCID: PMC4888473 DOI: 10.1186/s12943-016-0529-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/20/2016] [Indexed: 01/22/2023] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is an aggressive, locally invasive, cancer elicited by asbestos exposure and almost invariably a fatal diagnosis. To date, we are one of the leading laboratory that compared microRNA expression profiles in MPM and normal mesothelium samples in order to identify dysregulated microRNAs with functional roles in mesothelioma. We interrogated a significant collection of MPM tumors and normal pleural samples in our biobank in search for novel therapeutic targets. Methods Utilizing mRNA-microRNA correlations based on differential gene expression using Gene Set Enrichment Analysis (GSEA), we systematically combined publicly available gene expression datasets with our own MPM data in order to identify candidate targets for MPM therapy. Results We identified enrichment of target binding sites for the miR-17 and miR-30 families in both MPM tumors and cell lines. RT-qPCR revealed that members of both families were significantly downregulated in MPM tumors and cell lines. Interestingly, lower expression of miR-17-5p (P = 0.022) and miR-20a-5p (P = 0.026) was clearly associated with epithelioid histology. We interrogated the predicted targets of these differentially expressed microRNA families in MPM cell lines, and identified KCa1.1, a calcium-activated potassium channel subunit alpha 1 encoded by the KCNMA1 gene, as a target of miR-17-5p. KCa1.1 was overexpressed in MPM cells compared to the (normal) mesothelial line MeT-5A, and was also upregulated in patient tumor samples compared to normal mesothelium. Transfection of MPM cells with a miR-17-5p mimic or KCNMA1-specific siRNAs reduced mRNA expression of KCa1.1 and inhibited MPM cell migration. Similarly, treatment with paxilline, a small molecule inhibitor of KCa1.1, resulted in suppression of MPM cell migration. Conclusion These functional data implicating KCa1.1 in MPM cell migration support our integrative approach using MPM gene expression datasets to identify novel and potentially druggable targets. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0529-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuen Yee Cheng
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia
| | - Casey M Wright
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia
| | - Michaela B Kirschner
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia.,Division of Thoracic Surgery, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Marissa Williams
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia.,School of Medicine, University of Sydney, Sydney, NSW, 2006, Australia
| | - Kadir H Sarun
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Iryna Leshchynska
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - J James Edelman
- Cardiothoracic Surgical Unit, Royal Prince Alfred Hospital; The Baird Institute and Faculty of Medicine, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michael P Vallely
- Cardiothoracic Surgical Unit, Royal Prince Alfred Hospital; The Baird Institute and Faculty of Medicine, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Brian C McCaughan
- Sydney Cardiothoracic Surgeons, RPA Medical Centre, Sydney, NSW, 2050, Australia
| | - Sonja Klebe
- Department of Anatomical Pathology, Flinders Medical Centre, Adelaide, SA, 5042, Australia
| | - Nico van Zandwijk
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia.,School of Medicine, University of Sydney, Sydney, NSW, 2006, Australia
| | - Ruby C Y Lin
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia. .,School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Glen Reid
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia. .,School of Medicine, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
53
|
Zhou B, Sun Q, Kong DX. Predicting cancer-relevant proteins using an improved molecular similarity ensemble approach. Oncotarget 2016; 7:32394-407. [PMID: 27083051 PMCID: PMC5078021 DOI: 10.18632/oncotarget.8716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/28/2016] [Indexed: 12/15/2022] Open
Abstract
In this study, we proposed an improved algorithm for identifying proteins relevant to cancer. The algorithm was named two-layer molecular similarity ensemble approach (TL-SEA). We applied TL-SEA to analyzing the correlation between anticancer compounds (against cell lines K562, MCF7 and A549) and active compounds against separate target proteins listed in BindingDB. Several associations between cancer types and related proteins were revealed using this chemoinformatics approach. An analysis of the literature showed that 26 of 35 predicted proteins were correlated with cancer cell proliferation, apoptosis or differentiation. Additionally, interactions between proteins in BindingDB and anticancer chemicals were also predicted. We discuss the roles of the most important predicted proteins in cancer biology and conclude that TL-SEA could be a useful tool for inferring novel proteins involved in cancer and revealing underlying molecular mechanisms.
Collapse
Affiliation(s)
- Bin Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Qi Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - De-Xin Kong
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
54
|
Salem ML, Shoukry NM, Teleb WK, Abdel-Daim MM, Abdel-Rahman MA. In vitro and in vivo antitumor effects of the Egyptian scorpion Androctonus amoreuxi venom in an Ehrlich ascites tumor model. SPRINGERPLUS 2016; 5:570. [PMID: 27247867 PMCID: PMC4864766 DOI: 10.1186/s40064-016-2269-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/03/2016] [Indexed: 02/07/2023]
Abstract
Scorpion venom is a highly complex mixture of about 100-700 different components, where peptides are the major constituents with various biological and pharmacological properties including anticancer activities. In this study, anticancer efficacy of the venom of the Egyptian scorpion Androctonus amoreuxi has been evaluated. In vitro, the human breast cancer MCF-7 cell line was treated with the venom and the IC50 was estimated. In vivo studies, Ehrlich ascites carcinoma (EAC) cells were inoculated into CD-1 mice intraperitoneally to form liquid tumor or subcutaneously to form solid tumor and then treated with intraperitoneal injection with venom (0.22 mg/kg) every other day. The total tumor cells in the ascitic fluid and the size of the solid tumor were assessed after 14 and 30 days, respectively. In addition, the mean survival time (MST), body weight, tumor volume, PCV, viability of tumor cells, CBC, AST, ALP, creatinine, oxidative stress biomarkers (GSH, MDA, PCC), tumor marker Ki67, growth factor VEGF and caspase-3 were measured in normal control, EAC control and venom-treated groups (n = 6). Treatment with venom induced anti-tumor effects against liquid and in solid tumors as indicated by a significant (P < 0.05) reduction in tumor volume/size, count of viable EAC cells, expression of Ki67 and VEGF as well as by remarkable increases in MST and caspase-3 expression as compared to non-treated group. Interestingly, the venom restored the altered hematological and biochemical parameters of tumor-bearing animals and significantly increased their life span. These data indicate to (1) the cytotoxic potential effects of A. amoreuxi on tumor cells via anti-proliferative, apoptotic and anti-angiogenic activities; (2) opening a new avenue for further studies on the anti-cancer effects of this agent.
Collapse
Affiliation(s)
- Mohamed L. Salem
- />Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Nahla M. Shoukry
- />Zoology Department, Faculty of Science, Suez University, Suez, Egypt
| | - Wafaa K. Teleb
- />Zoology Department, Faculty of Science, Suez University, Suez, Egypt
| | - Mohamed M. Abdel-Daim
- />Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522 Egypt
| | | |
Collapse
|
55
|
Yu M, Liu SL, Sun PB, Pan H, Tian CL, Zhang LH. Peptide toxins and small-molecule blockers of BK channels. Acta Pharmacol Sin 2016; 37:56-66. [PMID: 26725735 DOI: 10.1038/aps.2015.139] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/13/2015] [Indexed: 12/21/2022] Open
Abstract
Large conductance, Ca(2+)-activated potassium (BK) channels play important roles in the regulation of neuronal excitability and the control of smooth muscle contractions. BK channels can be activated by changes in both the membrane potential and intracellular Ca(2+) concentrations. Here, we provide an overview of the structural and pharmacological properties of BK channel blockers. First, the properties of different venom peptide toxins from scorpions and snakes are described, with a focus on their characteristic structural motifs, including their disulfide bond formation pattern, the binding interface between the toxin and BK channel, and the functional consequence of the blockage of BK channels by these toxins. Then, some representative non-peptide blockers of BK channels are also described, including their molecular formula and pharmacological effects on BK channels. The detailed categorization and descriptions of these BK channel blockers will provide mechanistic insights into the blockade of BK channels. The structures of peptide toxins and non-peptide compounds could provide templates for the design of new channel blockers, and facilitate the optimization of lead compounds for further therapeutic applications in neurological disorders or cardiovascular diseases.
Collapse
|
56
|
Samuel P, Pink RC, Caley DP, Currie JMS, Brooks SA, Carter DRF. Over-expression of miR-31 or loss of KCNMA1 leads to increased cisplatin resistance in ovarian cancer cells. Tumour Biol 2015; 37:2565-73. [PMID: 26386726 DOI: 10.1007/s13277-015-4081-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/13/2015] [Indexed: 01/02/2023] Open
Abstract
Ovarian cancers have a high mortality rate; this is in part due to resistance to the platinum-based compounds used in chemotherapy. In this paper, we assess the role of microRNA-31 in the development of chemoresistance to cisplatin. We used previous data from microarray experiments to identify potential microRNAs (miRNAs) involved in chemoresistance. The functional significance of these microRNAs was tested using miRNA mimics. We used RNA-seq to identify pathways and genes de-regulated in the resistant cell line and then determined their role using RNAi. Analysis of publically available datasets reveals the potential clinical significance. Our data show that miR-31 is increased, whilst potassium channel calcium activated large conductance subfamily M alpha, member 1 (KCNMA1), a subunit of calcium-regulated big potassium (BK) channels, is reduced in resistant ovarian cells. Over-expression of miR-31 increased resistance, as did knockdown of KCNMA1 or inhibition of BK channels. This suggests that these genes directly modulate cisplatin response. Our data also suggest that miR-31 represses KCNMA1 expression. Comparing the levels of miR-31 and KCNMA1 to cisplatin resistance in the NCI60 panel or chemoresistance in cohorts of ovarian cancer tumours reveals correlations that support a role for these genes in vitro and in vivo. Here we show that miR-31 and KCNMA1 are involved in mediating cisplatin resistance in ovarian cancer. Our data gives a new insight into the potential mechanisms to therapeutically target in cisplatin resistance common to ovarian cancer.
Collapse
Affiliation(s)
- Priya Samuel
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
| | - Ryan Charles Pink
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
| | - Daniel Paul Caley
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK.,Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, V5Z 1L3, Canada
| | - James Michael Stevenson Currie
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
| | - Susan Ann Brooks
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK
| | - David Raul Francisco Carter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford, OX3 0BP, UK.
| |
Collapse
|
57
|
Hui L, Yang N, Yang H, Guo X, Jang X. Identification of biomarkers with a tumor stage-dependent expression and exploration of the mechanism involved in laryngeal squamous cell carcinoma. Oncol Rep 2015; 34:2627-35. [PMID: 26323359 DOI: 10.3892/or.2015.4230] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/02/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to identify biomarkers with a tumor stage-dependent expression manner and explore the regulatory mechanisms of laryngeal squamous cell carcinoma (LSCC) progression. Microarray data GSE59102 was used for differential analysis using a limma package. Enrichment analyses were performed for the differentially expressed genes (DEGs) between tumor tissues and normal tissues at different stages. A co-expressed network involving the overlapped DEGs in two stages was established based on Pearson's correlation coefficients. Furthermore, for the tumor stage‑dependent expressed DEGs, a protein‑protein interaction (PPI) network was constructed by mapping the genes using the STRING database. Transcription factors (TFs), oncogenes and tumor‑associated genes (TSGs) among the DEGs were predicted, following a search of the TRANSFAC, tumor-associated gene (TAG) and TSG databases. The CDT database was used to identify LSCC‑associated genes. In total, 696 DEGs from early stage and control samples and 622 DEGs from advanced sttage and control samples were selected, which were mainly enriched in the cell cycle pathway. In the co-expressed network, BUB1, TTK, E2F1 and CEP55 were prominent, with E2F1 being predicted as a TSG and CEP55 as an oncogene. The HOX family members were predicted as TFs. MMP1, MMP9, MMP3 and PLAU were the most evident nodes in the PPI network, where MMP3 was connected with MMP1. The ADH family was correlated with LSCC. Several biomarkers with tumor stage-dependent expression were identified including MMP1, MMP3, MMP9, PLAU and ADHs. Additionally, the dysregulated cell cycle pathway involving BUB1, TTK, E2F1 and CEP55, and the mediation of MMP1 by MMP3 as well as the predicted TF HOX, may all play significant roles in LSCC progression.
Collapse
Affiliation(s)
- Lian Hui
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ning Yang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Huijun Yang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xing Guo
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuejun Jang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
58
|
Morera FJ, Saravia J, Pontigo JP, Vargas-Chacoff L, Contreras GF, Pupo A, Lorenzo Y, Castillo K, Tilegenova C, Cuello LG, Gonzalez C. Voltage-dependent BK and Hv1 channels expressed in non-excitable tissues: New therapeutics opportunities as targets in human diseases. Pharmacol Res 2015; 101:56-64. [PMID: 26305431 DOI: 10.1016/j.phrs.2015.08.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/14/2015] [Accepted: 08/14/2015] [Indexed: 11/28/2022]
Abstract
Voltage-gated ion channels are the molecular determinants of cellular excitability. This group of ion channels is one of the most important pharmacological targets in excitable tissues such as nervous system, cardiac and skeletal muscle. Moreover, voltage-gated ion channels are expressed in non-excitable cells, where they mediate key cellular functions through intracellular biochemical mechanisms rather than rapid electrical signaling. This review aims at illustrating the pharmacological impact of these ion channels, highlighting in particular the structural details and physiological functions of two of them - the high conductance voltage- and Ca(2+)-gated K(+) (BK) channels and voltage-gated proton (Hv1) channels- in non-excitable cells. BK channels have been implicated in a variety of physiological processes ranging from regulation of smooth muscle tone to modulation of hormone and neurotransmitter release. Interestingly, BK channels are also involved in modulating K(+) transport in the mammalian kidney and colon epithelium with a potential role in the hyperkalemic phenotype observed in patients with familial hyperkalemic hypertension type 2, and in the pathophysiology of hypertension. In addition, BK channels are responsible for resting and stimulated Ca(2+)-activated K(+) secretion in the distal colon. Hv1 channels have been detected in many cell types, including macrophages, blood cells, lung epithelia, skeletal muscle and microglia. These channels have a central role in the phagocytic system. In macrophages, Hv1 channels participate in the generation of reactive oxygen species in the respiratory burst during the process of phagocytosis.
Collapse
Affiliation(s)
- Francisco J Morera
- Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile.
| | - Julia Saravia
- Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Juan Pablo Pontigo
- Institute of Marine Sciences and Limnology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Vargas-Chacoff
- Institute of Marine Sciences and Limnology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Gustavo F Contreras
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Amaury Pupo
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Yenisleidy Lorenzo
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Karen Castillo
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Cholpon Tilegenova
- Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubcock, TX, USA
| | - Luis G Cuello
- Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubcock, TX, USA.
| | - Carlos Gonzalez
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile.
| |
Collapse
|
59
|
Rabjerg M, Oliván-Viguera A, Hansen LK, Jensen L, Sevelsted-Møller L, Walter S, Jensen BL, Marcussen N, Köhler R. High expression of KCa3.1 in patients with clear cell renal carcinoma predicts high metastatic risk and poor survival. PLoS One 2015; 10:e0122992. [PMID: 25848765 PMCID: PMC4388734 DOI: 10.1371/journal.pone.0122992] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/26/2015] [Indexed: 01/08/2023] Open
Abstract
Background Ca2+-activated K+ channels have been implicated in cancer cell growth, metastasis, and tumor angiogenesis. Here we hypothesized that high mRNA and protein expression of the intermediate-conductance Ca2+-activated K+ channel, KCa3.1, is a molecular marker of clear cell Renal Cell Carcinoma (ccRCC) and metastatic potential and survival. Methodology/Principal Findings We analyzed channel expression by qRT-PCR, immunohistochemistry, and patch-clamp in ccRCC and benign oncocytoma specimens, in primary ccRCC and oncocytoma cell lines, as well as in two ccRCC cell lines (Caki-1 and Caki-2). CcRCC specimens contained 12-fold higher mRNA levels of KCa3.1 than oncocytoma specimens. The large-conductance channel, KCa1.1, was 3-fold more highly expressed in ccRCC than in oncocytoma. KCa3.1 mRNA expression in ccRCC was 2-fold higher than in the healthy cortex of the same kidney. Disease specific survival trended towards reduction in the subgroup of high-KCa3.1-expressing tumors (p<0.08 vs. low-KCa3.1-expressing tumors). Progression-free survival (time to metastasis/recurrence) was reduced significantly in the subgroup of high-KCa3.1-expressing tumors (p<0.02, vs. low-KCa3.1-expressing tumors). Immunohistochemistry revealed high protein expression of KCa3.1 in tumor vessels of ccRCC and oncocytoma and in a subset of ccRCC cells. Oncocytoma cells were devoid of KCa3.1 protein. In a primary ccRCC cell line and Caki-1/2-ccRCC cells, we found KCa3.1-protein as well as TRAM-34-sensitive KCa3.1-currents in a subset of cells. Furthermore, Caki-1/2-ccRCC cells displayed functional Paxilline-sensitive KCa1.1 currents. Neither KCa3.1 nor KCa1.1 were found in a primary oncocytoma cell line. Yet KCa-blockers, like TRAM-34 (KCa3.1) and Paxilline (KCa1.1), had no appreciable effects on Caki-1 proliferation in-vitro. Conclusions/Significance Our study demonstrated expression of KCa3.1 in ccRCC but not in benign oncocytoma. Moreover, high KCa3.1-mRNA expression levels were indicative of low disease specific survival of ccRCC patients, short progression-free survival, and a high metastatic potential. Therefore, KCa3.1 is of prognostic value in ccRCC.
Collapse
Affiliation(s)
- Maj Rabjerg
- Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
- * E-mail:
| | | | - Lars Koch Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Line Jensen
- Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Linda Sevelsted-Møller
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Steen Walter
- Department of Urology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Boye L. Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Niels Marcussen
- Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Ralf Köhler
- Aragon Institute of Health Sciences I+CS/IIS, 50009 Zaragoza, Spain
- Fundación Agencia Aragonesa para la Investigación y Desarrollo (ARAID), 50009 Zaragoza, Spain
| |
Collapse
|
60
|
Ortiz E, Gurrola GB, Schwartz EF, Possani LD. Scorpion venom components as potential candidates for drug development. Toxicon 2015; 93:125-35. [PMID: 25432067 PMCID: PMC7130864 DOI: 10.1016/j.toxicon.2014.11.233] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/25/2014] [Indexed: 10/25/2022]
Abstract
Scorpions are well known for their dangerous stings that can result in severe consequences for human beings, including death. Neurotoxins present in their venoms are responsible for their toxicity. Due to their medical relevance, toxins have been the driving force in the scorpion natural compounds research field. On the other hand, for thousands of years, scorpions and their venoms have been applied in traditional medicine, mainly in Asia and Africa. With the remarkable growth in the number of characterized scorpion venom components, several drug candidates have been found with the potential to tackle many of the emerging global medical threats. Scorpions have become a valuable source of biologically active molecules, from novel antibiotics to potential anticancer therapeutics. Other venom components have drawn attention as useful scaffolds for the development of drugs. This review summarizes the most promising candidates for drug development that have been isolated from scorpion venoms.
Collapse
Affiliation(s)
- Ernesto Ortiz
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autonóma de México, Avenida Universidad 2001, Cuernavaca 62210, Mexico
| | - Georgina B Gurrola
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autonóma de México, Avenida Universidad 2001, Cuernavaca 62210, Mexico
| | - Elisabeth Ferroni Schwartz
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília 70910-900, DF, Brazil
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autonóma de México, Avenida Universidad 2001, Cuernavaca 62210, Mexico.
| |
Collapse
|
61
|
Toro L, Li M, Zhang Z, Singh H, Wu Y, Stefani E. MaxiK channel and cell signalling. Pflugers Arch 2014; 466:875-86. [PMID: 24077696 DOI: 10.1007/s00424-013-1359-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/23/2023]
Abstract
The large-conductance Ca2+- and voltage-activated K+ (MaxiK, BK, BKCa, Slo1, KCa1.1) channel role in cell signalling is becoming apparent as we learn how the channel interacts with a multiplicity of proteins not only at the plasma membrane but also in intracellular organelles including the endoplasmic reticulum, nucleus, and mitochondria. In this review, we focus on the interactions of MaxiK channels with seven-transmembrane G protein-coupled receptors and discuss information suggesting that, the channel big C-terminus may act as the nucleus of signalling molecules including kinases relevant for cell death and survival. Increasing evidence indicates that the channel is able to associate with a variety of receptors including β-adrenergic receptors, G protein-coupled estrogen receptors, acetylcholine receptors, thromboxane A2 receptors, and angiotensin II receptors, which highlights the varied functions that the channel has (or may have) not only in regulating contraction/relaxation of muscle cells or neurotransmission in the brain but also in cell metabolism, proliferation, migration, and gene expression. In line with this view, MaxiK channels have been implicated in obesity and in brain, prostate, and mammary cancers. A better understanding on the molecular mechanisms underlying or triggered by MaxiK channel abnormalities like overexpression in certain cancers may lead to new therapeutics to prevent devastating diseases.
Collapse
|
62
|
Ion channel expression as promising cancer biomarker. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2685-702. [PMID: 25542783 DOI: 10.1016/j.bbamem.2014.12.016] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/10/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022]
Abstract
Cancer is a disease with marked heterogeneity in both response to therapy and survival. Clinical and histopathological characteristics have long determined prognosis and therapy. The introduction of molecular diagnostics has heralded an explosion in new prognostic factors. Overall, histopathology, immunohistochemistry and molecular biology techniques have described important new prognostic subgroups in the different cancer categories. Ion channels and transporters (ICT) are a new class of membrane proteins which are aberrantly expressed in several types of human cancers. Besides regulating different aspect of cancer cell behavior, ICT can now represent novel cancer biomarkers. A summary of the data obtained so far and relative to breast, prostate, lung, colorectal, esophagus, pancreatic and gastric cancers are reported. Special emphasis is given to those studies aimed at relating specific ICT or a peculiar ICT profile with current diagnostic methods. Overall, we are close to exploit ICTs for diagnostic, prognostic or predictive purposes in cancer. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
|
63
|
Curci A, Mele A, Camerino GM, Dinardo MM, Tricarico D. The large conductance Ca(2+) -activated K(+) (BKCa) channel regulates cell proliferation in SH-SY5Y neuroblastoma cells by activating the staurosporine-sensitive protein kinases. Front Physiol 2014; 5:476. [PMID: 25538629 PMCID: PMC4260485 DOI: 10.3389/fphys.2014.00476] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/20/2014] [Indexed: 12/31/2022] Open
Abstract
Here we investigated on the role of the calcium activated K(+)-channels(BKCa) on the regulation of the neuronal viability. Recordings of the K(+)-channel current were performed using patch-clamp technique in human neuroblastoma cells (SH-SY5Y) in parallel with measurements of the cell viability in the absence or presence of the BKCa channel blockers iberiotoxin(IbTX) and tetraethylammonium (TEA) and the BKCa channel opener NS1619. Protein kinase C/A (PKC, PKA) activities in the cell lysate were investigated in the presence/absence of drugs. The whole-cell K(+)-current showed a slope conductance calculated at negative membrane potentials of 126.3 pS and 1.717 nS(n = 46) following depolarization. The intercept of the I/V curve was -33 mV. IbTX(10(-8) - 4 × 10(-7) M) reduced the K(+)-current at +30 mV with an IC50 of 1.85 × 10(-7) M and an Imax of -46% (slope = 2.198) (n = 21). NS1619(10-100 × 10(-6) M) enhanced the K(+)-current of +141% (n = 6), at -10 mV(Vm). TEA(10(-5)-10(-3) M) reduced the K(+)-current with an IC50 of 3.54 × 10(-5) M and an Imax of -90% (slope = 0.95) (n = 5). A concentration-dependent increase of cell proliferation was observed with TEA showing a maximal proliferative effect(MPE) of +38% (10(-4) M). IbTX showed an MPE of +42% at 10(-8) M concentration, reducing it at higher concentrations. The MPE of the NS1619(100 × 10(-6) M) was +42%. The PKC inhibitor staurosporine (0.2-2 × 10(-6) M) antagonized the proliferative actions of IbTX and TEA. IbTX (10 × 10(-9) M), TEA (100 × 10(-6) M), and the NS1619 significantly enhanced the PKC and PKA activities in the cell lysate with respect to the controls. These results suggest that BKCa channel regulates proliferation of the SH-SY5Y cells through PKC and PKA protein kinases.
Collapse
Affiliation(s)
- Angela Curci
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro" Bari, Italy
| | - Antonietta Mele
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro" Bari, Italy
| | | | | | - Domenico Tricarico
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro" Bari, Italy
| |
Collapse
|
64
|
Schickling BM, England SK, Aykin-Burns N, Norian LA, Leslie KK, Frieden-Korovkina VP. BKCa channel inhibitor modulates the tumorigenic ability of hormone-independent breast cancer cells via the Wnt pathway. Oncol Rep 2014; 33:533-8. [PMID: 25422049 PMCID: PMC4306270 DOI: 10.3892/or.2014.3617] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/12/2014] [Indexed: 11/05/2022] Open
Abstract
In breast cancers, the large conductance Ca2+ and voltage sensitive K+ (BKCa) channels have been hypothesized to function as oncoproteins, yet it remains unclear how inhibition of channel activity impacts oncogenesis. We demonstrated herein that iberiotoxin (IbTX), an inhibitor of BKCa channels, differentially modulated the in vitro tumorigenic activities of hormone-independent breast cancer cells. Specifically, in HER-2/neu-overexpressing UACC893 cells and triple‑negative MDA-MB-231 cells, IbTX selectively attenuated anchorage-independent growth with concomitant downregulation of β-catenin as well as total and phosphorylated Akt and HER-2/neu. By contrast, HER-2/neu-overexpressing SK-BR-3 cells were insensitive to IbTX. Molecular analyses showed an absence of β-catenin and a dose-dependent upregulation of total and phosphorylated Akt and HER-2/neu in these cells. Taken together, these studies identify β-catenin as a putative modulator of the inhibitory actions of IbTX in sensitive breast cancer cells.
Collapse
Affiliation(s)
| | - Sarah K England
- Division of Basic Science Research, Washington University, St. Louis, MO, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lyse A Norian
- Department of Urology, University of Iowa, Iowa City, IA, USA
| | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
65
|
Du C, Chen L, Zhang H, Wang Z, Liu W, Xie X, Xie M. Caveolin-1 limits the contribution of BKCa channel to MCF-7 breast cancer cell proliferation and invasion. Int J Mol Sci 2014; 15:20706-22. [PMID: 25397596 PMCID: PMC4264191 DOI: 10.3390/ijms151120706] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/09/2014] [Accepted: 10/22/2014] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence suggests that caveolin-1 and large conductance Ca2+-activated potassium (BKCa) channels are implicated in the carcinogenesis processes, including cell proliferation and invasion. These two proteins have been proven to interact with each other in vascular endothelial and smooth muscle cells and modulate vascular contractility. In this study, we investigated the probable interaction between caveolin-1 and BKCa in MCF-7 breast cancer cells. We identified that caveolin-1 and BKCa were co-localized and could be reciprocally co-immunoprecipitated in human breast cancer MCF-7 cells. siRNA mediated caveolin-1 knockdown resulted in activation and increased surface expression of BKCa channel, and subsequently promoted the proliferation and invasiveness of breast cancer cells. These effects were attenuated in the presence of BKCa-siRNA. Conversely, up-regulated caveolin-1 suppressed function and surface expression of BKCa channel and exerted negative effects on breast cancer cell proliferation and invasion. Similarly, these opposing effects were abrogated by BKCa up-regulation. Collectively, our findings suggest that BKCa is a critical target for suppression by caveolin-1 in suppressing proliferation and invasion of breast cancer cells. The functional complex of caveolin-1 and BKCa in the membrane microdomain may be served as a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Cheng Du
- Department of Oncology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China.
| | - Li Chen
- Key Laboratory of Aerospace Medicine, Ministry of Education, the Fourth Military Medical University, Xi'an 710032, China.
| | - Haijun Zhang
- Key Laboratory of Aerospace Medicine, Ministry of Education, the Fourth Military Medical University, Xi'an 710032, China.
| | - Zhongchao Wang
- Key Laboratory of Aerospace Medicine, Ministry of Education, the Fourth Military Medical University, Xi'an 710032, China.
| | - Wenchao Liu
- Department of Oncology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China.
| | - Xiaodong Xie
- Department of Oncology, General Hospital of Shenyang Military Area Command, Shenyang 110840, China.
| | - Manjiang Xie
- Key Laboratory of Aerospace Medicine, Ministry of Education, the Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
66
|
Ge L, Hoa NT, Wilson Z, Arismendi-Morillo G, Kong XT, Tajhya RB, Beeton C, Jadus MR. Big Potassium (BK) ion channels in biology, disease and possible targets for cancer immunotherapy. Int Immunopharmacol 2014; 22:427-43. [PMID: 25027630 PMCID: PMC5472047 DOI: 10.1016/j.intimp.2014.06.040] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 06/27/2014] [Accepted: 06/30/2014] [Indexed: 11/18/2022]
Abstract
The Big Potassium (BK) ion channel is commonly known by a variety of names (Maxi-K, KCNMA1, slo, stretch-activated potassium channel, KCa1.1). Each name reflects a different physical property displayed by this single ion channel. This transmembrane channel is found on nearly every cell type of the body and has its own distinctive roles for that tissue type. The BKα channel contains the pore that releases potassium ions from intracellular stores. This ion channel is found on the cell membrane, endoplasmic reticulum, Golgi and mitochondria. Complex splicing pathways produce different isoforms. The BKα channels can be phosphorylated, palmitoylated and myristylated. BK is composed of a homo-tetramer that interacts with β and γ chains. These accessory proteins provide a further modulating effect on the functions of BKα channels. BK channels play important roles in cell division and migration. In this review, we will focus on the biology of the BK channel, especially its role, and its immune response towards cancer. Recent proteomic studies have linked BK channels with various proteins. Some of these interactions offer further insight into the role that BK channels have with cancers, especially with brain tumors. This review shows that BK channels have a complex interplay with intracellular components of cancer cells and still have plenty of secrets to be discovered.
Collapse
Affiliation(s)
- Lisheng Ge
- Research Service, VA Long Beach Healthcare System, 5901 E. 7th Street, Long Beach, CA 90822, USA
| | - Neil T Hoa
- Research Service, VA Long Beach Healthcare System, 5901 E. 7th Street, Long Beach, CA 90822, USA
| | - Zechariah Wilson
- Research Service, VA Long Beach Healthcare System, 5901 E. 7th Street, Long Beach, CA 90822, USA
| | | | - Xiao-Tang Kong
- Department of Neuro-Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rajeev B Tajhya
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Martin R Jadus
- Research Service, VA Long Beach Healthcare System, 5901 E. 7th Street, Long Beach, CA 90822, USA; Pathology and Laboratory Medicine Service, VA Long Beach Healthcare System, 5901 E. 7th Street, Long Beach, CA 90822, USA; Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of California, Irvine, Orange, CA 92868, USA; Pathology and Laboratory Medicine, Med Sci I, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
67
|
Ding J, Chua PJ, Bay BH, Gopalakrishnakone P. Scorpion venoms as a potential source of novel cancer therapeutic compounds. Exp Biol Med (Maywood) 2014; 239:387-93. [PMID: 24599885 DOI: 10.1177/1535370213513991] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Scorpions and their venoms have been used in traditional medicine for thousands of years in China, India and Africa. The scorpion venom is a highly complex mixture of salts, nucleotides, biogenic amines, enzymes, mucoproteins, as well as peptides and proteins (e.g. neurotoxins). One of the recently observed biological properties of animal venoms and toxins is that they possess anticancer potential. An increasing number of studies have shown that scorpion venoms and toxins can decrease cancer growth, induce apoptosis and inhibit cancer progression and metastasis in vitro and in vivo. Several active molecules with anticancer activities, ranging from inhibition of proliferation and cell cycle arrest to induction of apoptosis and decreasing cell migration and invasion, have been isolated from scorpion venoms. These observations have shed light on the application of scorpion venoms and toxins as potential novel cancer therapeutics. This mini-review focuses on the anticancer potential of scorpion venoms and toxins and the possible mechanisms for their antitumor activities.
Collapse
Affiliation(s)
- Jian Ding
- Venom and Toxin Research Programme, Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597
| | | | | | | |
Collapse
|
68
|
Effects of Ca2+-activated potassium and inward rectifier potassium channel on the differentiation of endothelial progenitor cells from human peripheral blood. Mol Biol Rep 2014; 41:3413-23. [DOI: 10.1007/s11033-014-3203-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 01/25/2014] [Indexed: 11/26/2022]
|
69
|
Identification of candidate driver genes in common focal chromosomal aberrations of microsatellite stable colorectal cancer. PLoS One 2013; 8:e83859. [PMID: 24367615 PMCID: PMC3867468 DOI: 10.1371/journal.pone.0083859] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/08/2013] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer deaths worldwide. Chromosomal instability (CIN) is a major driving force of microsatellite stable (MSS) sporadic CRC. CIN tumours are characterised by a large number of somatic chromosomal copy number aberrations (SCNA) that frequently affect oncogenes and tumour suppressor genes. The main aim of this work was to identify novel candidate CRC driver genes affected by recurrent and focal SCNA. High resolution genome-wide comparative genome hybridisation (CGH) arrays were used to compare tumour and normal DNA for 53 sporadic CRC cases. Context corrected common aberration (COCA) analysis and custom algorithms identified 64 deletions and 32 gains of focal minimal common regions (FMCR) at high frequency (>10%). Comparison of these FMCR with published genomic profiles from CRC revealed common overlap (42.2% of deletions and 34.4% of copy gains). Pathway analysis showed that apoptosis and p53 signalling pathways were commonly affected by deleted FMCR, and MAPK and potassium channel pathways by gains of FMCR. Candidate tumour suppressor genes in deleted FMCR included RASSF3, IFNAR1, IFNAR2 and NFKBIA and candidate oncogenes in gained FMCR included PRDM16, TNS1, RPA3 and KCNMA1. In conclusion, this study confirms some previously identified aberrations in MSS CRC and provides in silico evidence for some novel candidate driver genes.
Collapse
|
70
|
Contreras GF, Castillo K, Enrique N, Carrasquel-Ursulaez W, Castillo JP, Milesi V, Neely A, Alvarez O, Ferreira G, González C, Latorre R. A BK (Slo1) channel journey from molecule to physiology. Channels (Austin) 2013; 7:442-58. [PMID: 24025517 DOI: 10.4161/chan.26242] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium and voltage-activated potassium (BK) channels are key actors in cell physiology, both in neuronal and non-neuronal cells and tissues. Through negative feedback between intracellular Ca (2+) and membrane voltage, BK channels provide a damping mechanism for excitatory signals. Molecular modulation of these channels by alternative splicing, auxiliary subunits and post-translational modifications showed that these channels are subjected to many mechanisms that add diversity to the BK channel α subunit gene. This complexity of interactions modulates BK channel gating, modifying the energetic barrier of voltage sensor domain activation and channel opening. Regions for voltage as well as Ca (2+) sensitivity have been identified, and the crystal structure generated by the 2 RCK domains contained in the C-terminal of the channel has been described. The linkage of these channels to many intracellular metabolites and pathways, as well as their modulation by extracellular natural agents, has been found to be relevant in many physiological processes. This review includes the hallmarks of BK channel biophysics and its physiological impact on specific cells and tissues, highlighting its relationship with auxiliary subunit expression.
Collapse
Affiliation(s)
- Gustavo F Contreras
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile; Doctorado en Ciencias mención Neurociencia; Universidad de Valparaíso; Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| | - Nicolás Enrique
- Grupo de Investigación en Fisiología Vascular (GINFIV); Universidad Nacional de la Plata; La Plata, Argentina
| | - Willy Carrasquel-Ursulaez
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile; Doctorado en Ciencias mención Neurociencia; Universidad de Valparaíso; Valparaíso, Chile
| | - Juan Pablo Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile; Facultad de Ciencias; Universidad de Chile; Santiago, Chile
| | - Verónica Milesi
- Grupo de Investigación en Fisiología Vascular (GINFIV); Universidad Nacional de la Plata; La Plata, Argentina
| | - Alan Neely
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| | | | - Gonzalo Ferreira
- Laboratorio de Canales Iónicos; Departamento de Biofísica; Facultad de Medicina; Universidad de la República; Montevideo, Uruguay
| | - Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso; Facultad de Ciencias; Universidad de Valparaíso; Valparaíso, Chile
| |
Collapse
|
71
|
Gackière F, Warnier M, Katsogiannou M, Derouiche S, Delcourt P, Dewailly E, Slomianny C, Humez S, Prevarskaya N, Roudbaraki M, Mariot P. Functional coupling between large-conductance potassium channels and Cav3.2 voltage-dependent calcium channels participates in prostate cancer cell growth. Biol Open 2013; 2:941-51. [PMID: 24143281 PMCID: PMC3773341 DOI: 10.1242/bio.20135215] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 06/26/2013] [Indexed: 12/11/2022] Open
Abstract
It is strongly suspected that potassium (K+) channels are involved in various aspects of prostate cancer development, such as cell growth. However, the molecular nature of those K+ channels implicated in prostate cancer cell proliferation and the mechanisms through which they control proliferation are still unknown. This study uses pharmacological, biophysical and molecular approaches to show that the main voltage-dependent K+ current in prostate cancer LNCaP cells is carried by large-conductance BK channels. Indeed, most of the voltage-dependent current was inhibited by inhibitors of BK channels (paxillin and iberiotoxin) and by siRNA targeting BK channels. In addition, we reveal that BK channels constitute the main K+ channel family involved in setting the resting membrane potential in LNCaP cells at around −40 mV. This consequently promotes a constitutive calcium entry through T-type Cav3.2 calcium channels. We demonstrate, using single-channel recording, confocal imaging and co-immunoprecipitation approaches, that both channels form macromolecular complexes. Finally, using flow cytometry cell cycle measurements, cell survival assays and Ki67 immunofluorescent staining, we show that both BK and Cav3.2 channels participate in the proliferation of prostate cancer cells.
Collapse
Affiliation(s)
- Florian Gackière
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Bâtiment SN3, Université Lille 1 , 59655 Villeneuve d'Ascq Cédex , France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Haddad BR, Gu L, Mirtti T, Dagvadorj A, Vogiatzi P, Hoang DT, Bajaj R, Leiby B, Ellsworth E, Blackmon S, Ruiz C, Curtis M, Fortina P, Ertel A, Liu C, Rui H, Visakorpi T, Bubendorf L, Lallas CD, Trabulsi EJ, McCue P, Gomella L, Nevalainen MT. STAT5A/B gene locus undergoes amplification during human prostate cancer progression. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:2264-75. [PMID: 23660011 DOI: 10.1016/j.ajpath.2013.02.044] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 02/22/2013] [Accepted: 02/28/2013] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms underlying progression of prostate cancer (PCa) to castrate-resistant (CR) and metastatic disease are poorly understood. Our previous mechanistic work shows that inhibition of transcription factor Stat5 by multiple alternative methods induces extensive rapid apoptotic death of Stat5-positive PCa cells in vitro and inhibits PCa xenograft tumor growth in nude mice. Furthermore, STAT5A/B induces invasive behavior of PCa cells in vitro and in vivo, suggesting involvement of STAT5A/B in PCa progression. Nuclear STAT5A/B protein levels are increased in high-grade PCas, CR PCas, and distant metastases, and high nuclear STAT5A/B expression predicts early disease recurrence and PCa-specific death in clinical PCas. Based on these findings, STAT5A/B represents a therapeutic target protein for advanced PCa. The mechanisms underlying increased Stat5 protein levels in PCa are unclear. Herein, we demonstrate amplification at the STAT5A/B gene locus in a significant fraction of clinical PCa specimens. STAT5A/B gene amplification was more frequently found in PCas of high histologic grades and in CR distant metastases. Quantitative in situ analysis revealed that STAT5A/B gene amplification was associated with increased STAT5A/B protein expression in PCa. Functional studies showed that increased STAT5A/B copy numbers conferred growth advantage in PCa cells in vitro and as xenograft tumors in vivo. The work presented herein provides the first evidence of somatic STAT5A/B gene amplification in clinical PCas.
Collapse
Affiliation(s)
- Bassem R Haddad
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
State-dependent FRET reports calcium- and voltage-dependent gating-ring motions in BK channels. Proc Natl Acad Sci U S A 2013; 110:5217-22. [PMID: 23479636 DOI: 10.1073/pnas.1219611110] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Large-conductance voltage- and calcium-dependent potassium channels (BK, "Big K+") are important controllers of cell excitability. In the BK channel, a large C-terminal intracellular region containing a "gating-ring" structure has been proposed to transduce Ca(2+) binding into channel opening. Using patch-clamp fluorometry, we have investigated the calcium and voltage dependence of conformational changes of the gating-ring region of BK channels, while simultaneously monitoring channel conductance. Fluorescence resonance energy transfer (FRET) between fluorescent protein inserts indicates that Ca(2+) binding produces structural changes of the gating ring that are much larger than those predicted by current X-ray crystal structures of isolated gating rings.
Collapse
|
74
|
Single Mechanosensitive and Ca2+-Sensitive Channel Currents Recorded from Mouse and Human Embryonic Stem Cells. J Membr Biol 2012. [DOI: 10.1007/s00232-012-9523-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
75
|
Ruiz C, Martins JR, Rudin F, Schneider S, Dietsche T, Fischer CA, Tornillo L, Terracciano LM, Schreiber R, Bubendorf L, Kunzelmann K. Enhanced expression of ANO1 in head and neck squamous cell carcinoma causes cell migration and correlates with poor prognosis. PLoS One 2012; 7:e43265. [PMID: 22912841 PMCID: PMC3422276 DOI: 10.1371/journal.pone.0043265] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/18/2012] [Indexed: 12/23/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has the potential for early metastasis and is associated with poor survival. Ano1 (Dog1) is an established and sensitive marker for the diagnosis of gastrointestinal stromal tumors (GIST) and has recently been identified as a Ca2+ activated Cl− channel. Although the ANO1 gene is located on the 11q13 locus, a region which is known to be amplified in different types of human carcinomas, a detailed analysis of Ano1 amplification and expression in HNSCC has not been performed. It is thus still unclear how Ano1 contributes to malignancy in HNSCC. We analyzed genomic amplification of the 11q13 locus and Ano1 together with Ano1-protein expression in a large collection of HNSCC samples. We detected a highly significant correlation between amplification and expression of Ano1 and showed that HNSCC patients with Ano1 protein expression have a poor overall survival. We further analyzed the expression of the Ano1 protein in more than 4′000 human samples from 80 different tumor types and 76 normal tissue types and detected that besides HNSCC and GISTs, Ano1 was rarely expressed in other tumor samples or healthy human tissues. In HNSCC cell lines, expression of Ano1 caused Ca2+ activated Cl− currents, which induced cell motility and cell migration in wound healing and in real time migration assays, respectively. In contrast, knockdown of Ano1 did not affect intracellular Ca2+ signaling and surprisingly did not reduce cell proliferation in BHY cells. Further, expression and activity of Ano1 strongly correlated with the ability of HNSCC cells to regulate their volume. Thus, poor survival in HNSCC patients is correlated with the presence of Ano1. Our results further suggest that Ano1 facilitates regulation of the cell volume and causes cell migration, which both can contribute to metastatic progression in HNSCC.
Collapse
Affiliation(s)
- Christian Ruiz
- Institute for Pathology, University Hospital Basel, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Abstract
KCNMA1 encodes the α-subunit of the large conductance, voltage and Ca2+-activated (BK) potassium channel and has been reported as a target gene of genomic amplification at 10q22 in prostate cancer. To investigate the prevalence of the amplification in other human cancers, the copy number of KCNMA1 was analyzed by fluorescence-in-situ-hybridization (FISH) in 2,445 tumors across 118 different tumor types. Amplification of KCNMA1 was restricted to a small but distinct fraction of breast, ovarian and endometrial cancer with the highest prevalence in invasive ductal breast cancers and serous carcinoma of ovary and endometrium (3–7%). We performed an extensive analysis on breast cancer tissue microarrays (TMA) of 1,200 tumors linked to prognosis. KCNMA1 amplification was significantly associated with high tumor stage, high grade, high tumor cell proliferation, and poor prognosis. Immunofluorescence revealed moderate or strong KCNMA1 protein expression in 8 out of 9 human breast cancers and in the breast cancer cell line MFM223. KCNMA1-function in breast cancer cell lines was confirmed by whole-cell patch clamp recordings and proliferation assays, using siRNA-knockdown, BK channel activators such as 17ß-estradiol and the BK-channel blocker paxilline. Our findings revealed that enhanced expression of KCNMA1 correlates with and contributes to high proliferation rate and malignancy of breast cancer.
Collapse
|
77
|
Huang X, Dubuc AM, Hashizume R, Berg J, He Y, Wang J, Chiang C, Cooper MK, Northcott PA, Taylor MD, Barnes MJ, Tihan T, Chen J, Hackett CS, Weiss WA, James CD, Rowitch DH, Shuman MA, Jan YN, Jan LY. Voltage-gated potassium channel EAG2 controls mitotic entry and tumor growth in medulloblastoma via regulating cell volume dynamics. Genes Dev 2012; 26:1780-96. [PMID: 22855790 DOI: 10.1101/gad.193789.112] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.
Collapse
Affiliation(s)
- Xi Huang
- Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Ge L, Hoa NT, Cornforth AN, Bota DA, Mai A, Kim DI, Chiou SK, Hickey MJ, Kruse CA, Jadus MR. Glioma big potassium channel expression in human cancers and possible T cell epitopes for their immunotherapy. THE JOURNAL OF IMMUNOLOGY 2012; 189:2625-34. [PMID: 22844111 DOI: 10.4049/jimmunol.1102965] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Big potassium (BK) ion channels have several spliced variants. One spliced variant initially described within human glioma cells is the glioma BK (gBK) channel. This isoform consists of 34 aa inserted into the intracellular region of the basic BK ion channel. PCR primers specific for this inserted region confirmed that human glioma cell lines and freshly resected surgical tissues from glioblastoma multiforme patients strongly expressed gBK mRNA. Normal human brain tissue very weakly expressed this transcript. An Ab specific for this gBK isoform confirmed that human glioma cells displayed this protein in the cell membrane, mitochondria, Golgi, and endoplasmic reticulum. Within the gBK region, two putative epitopes (gBK1 and gBK2) are predicted to bind to the HLA-A*0201 molecule. HLA-A*0201-restricted human CTLs were generated in vitro using gBK peptide-pulsed dendritic cells. Both gBK1 and gBK2 peptide-specific CTLs killed HLA-A2⁺/gBK⁺ gliomas, but they failed to kill non-HLA-A2-expressing but gBK⁺ target cells in cytolytic assays. T2 cells loaded with exogenous gBK peptides, but not with the influenza M1 control peptide, were only killed by their respective CTLs. The gBK-specific CTLs also killed a variety of other HLA-A*0201⁺ cancer cells that possess gBK, as well as HLA-A2⁺ HEK cells transfected with the gBK gene. Of clinical relevance, we found that T cells derived from glioblastoma multiforme patients that were sensitized to the gBK peptide could also kill target cells expressing gBK. This study shows that peptides derived from cancer-associated ion channels maybe useful targets for T cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Lisheng Ge
- Diagnostic and Molecular Health Care Group, Long Beach, CA 90822, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Ma YG, Liu WC, Dong S, Du C, Wang XJ, Li JS, Xie XP, Wu L, Ma DC, Yu ZB, Xie MJ. Activation of BK(Ca) channels in zoledronic acid-induced apoptosis of MDA-MB-231 breast cancer cells. PLoS One 2012; 7:e37451. [PMID: 22655048 PMCID: PMC3360057 DOI: 10.1371/journal.pone.0037451] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 04/20/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Zoledronic acid, one of the most potent nitrogen-containing biphosphonates, has been demonstrated to have direct anti-tumor and anti-metastatic properties in breast cancer in vitro and in vivo. In particular, tumor-cell apoptosis has been recognized to play an important role in the treatment of metastatic breast cancer with zoledronic acid. However, the precise mechanisms remain less clear. In the present study, we investigated the specific role of large conductance Ca(2+)-activated potassium (BK(Ca)) channel in zoledronic acid-induced apoptosis of estrogen receptor (ER)-negative MDA-MB-231 breast cancer cells. METHODOLOGY/PRINCIPAL FINDINGS The action of zoledronic acid on BK(Ca) channel was investigated by whole-cell and cell-attached patch clamp techniques. Cell apoptosis was assessed with immunocytochemistry, analysis of fragmented DNA by agarose gel electrophoresis, and flow cytometry assays. Cell proliferation was investigated by MTT test and immunocytochemistry. In addition, such findings were further confirmed with human embryonic kidney 293 (HEK293) cells which were transfected with functional BK(Ca) α-subunit (hSloα). Our results clearly indicated that zoledronic acid directly increased the activities of BK(Ca) channels, and then activation of BK(Ca) channel by zoledronic acid contributed to induce apoptosis in MDA-MB-231 cells. The possible mechanisms were associated with the elevated level of intracellular Ca(2+) and a concomitant depolarization of mitochondrial membrane potential (Δψm) in MDA-MB-231 cells. CONCLUSIONS Activation of BK(Ca) channel was here shown to be a novel molecular pathway involved in zoledronic acid-induced apoptosis of MDA-MB-231 cells in vitro.
Collapse
Affiliation(s)
- Yu-Guang Ma
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Department of Breast Disease, First Hospital of Lanzhou University, Lanzhou, China
| | - Wen-Chao Liu
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- * E-mail: (WCL); (MJX)
| | - Shuo Dong
- Department of Medicine, Baylor College of Medicine, Houston, United States of America
| | - Cheng Du
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiao-Jun Wang
- Key Laboratory of Aerospace Medicine, Department of Aerospace Physiology, Fourth Military Medical University, Ministry of China, Xi'an, China
| | - Jin-Sheng Li
- Key Laboratory of Aerospace Medicine, Department of Aerospace Physiology, Fourth Military Medical University, Ministry of China, Xi'an, China
| | - Xiao-Ping Xie
- Key Laboratory of Aerospace Medicine, Department of Aerospace Physiology, Fourth Military Medical University, Ministry of China, Xi'an, China
| | - Li Wu
- Department of Breast Disease, First Hospital of Lanzhou University, Lanzhou, China
| | - Da-Chang Ma
- Department of Breast Disease, First Hospital of Lanzhou University, Lanzhou, China
| | - Zhi-Bin Yu
- Key Laboratory of Aerospace Medicine, Department of Aerospace Physiology, Fourth Military Medical University, Ministry of China, Xi'an, China
| | - Man-Jiang Xie
- Key Laboratory of Aerospace Medicine, Department of Aerospace Physiology, Fourth Military Medical University, Ministry of China, Xi'an, China
- * E-mail: (WCL); (MJX)
| |
Collapse
|
80
|
Large conductance, calcium- and voltage-gated potassium (BK) channels: regulation by cholesterol. Pharmacol Ther 2012; 135:133-50. [PMID: 22584144 DOI: 10.1016/j.pharmthera.2012.05.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 04/09/2012] [Indexed: 11/21/2022]
Abstract
Cholesterol (CLR) is an essential component of eukaryotic plasma membranes. CLR regulates the membrane physical state, microdomain formation and the activity of membrane-spanning proteins, including ion channels. Large conductance, voltage- and Ca²⁺-gated K⁺ (BK) channels link membrane potential to cell Ca²⁺ homeostasis. Thus, they control many physiological processes and participate in pathophysiological mechanisms leading to human disease. Because plasmalemma BK channels cluster in CLR-rich membrane microdomains, a major driving force for studying BK channel-CLR interactions is determining how membrane CLR controls the BK current phenotype, including its pharmacology, channel sorting, distribution, and role in cell physiology. Since both BK channels and CLR tissue levels play a pathophysiological role in human disease, identifying functional and structural aspects of the CLR-BK channel interaction may open new avenues for therapeutic intervention. Here, we review the studies documenting membrane CLR-BK channel interactions, dissecting out the many factors that determine the final BK current response to changes in membrane CLR content. We also summarize work in reductionist systems where recombinant BK protein is studied in artificial lipid bilayers, which documents a direct inhibition of BK channel activity by CLR and builds a strong case for a direct interaction between CLR and the BK channel-forming protein. Bilayer lipid-mediated mechanisms in CLR action are also discussed. Finally, we review studies of BK channel function during hypercholesterolemia, and underscore the many consequences that the CLR-BK channel interaction brings to cell physiology and human disease.
Collapse
|
81
|
Kim Y, Kim WJ, Cha EJ. Quercetin-induced Growth Inhibition in Human Bladder Cancer Cells Is Associated with an Increase in Ca-activated K Channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2011; 15:279-83. [PMID: 22128260 DOI: 10.4196/kjpp.2011.15.5.279] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/03/2011] [Accepted: 10/09/2011] [Indexed: 12/25/2022]
Abstract
Quercetin (3,3',4',5,7-pentahydroxyflavone) is an attractive therapeutic flavonoid for cancer treatment because of its beneficial properties including apoptotic, antioxidant, and antiproliferative effects on cancer cells. However, the exact mechanism of action of quercetin on ion channel modulation is poorly understood in bladder cancer 253J cells. In this study, we demonstrated that large conductance Ca(2+)-activated K(+) (BK(Ca)) or MaxiK channels were functionally expressed in 253J cells, and quercetin increased BK(Ca) current in a concentration dependent and reversible manner using a whole cell patch configuration. The half maximal activation concentration (IC(50)) of quercetin was 45.5±7.2 µM. The quercetin-evoked BK(Ca) current was inhibited by tetraethylammonium (TEA; 5 mM) a non-specific BK(Ca) blocker and iberiotoxin (IBX; 100 nM) a BK(Ca)-specific blocker. Quercetin-induced membrane hyperpolarization was measured by fluorescence-activated cell sorting (FACS) with voltage sensitive dye, bis (1,3-dibutylbarbituric acid) trimethine oxonol (DiBAC(4)(3); 100 nM). Quercetin-evoked hyperpolarization was prevented by TEA. Quercetin produced an antiproliferative effect (30.3±13.5%) which was recovered to 53.3±10.5% and 72.9±3.7% by TEA and IBX, respectively. Taken together our results indicate that activation of BK(Ca) channels may be considered an important target related to the action of quercetin on human bladder cancer cells.
Collapse
Affiliation(s)
- Yangmi Kim
- Department of Physiology, Chungbuk National University, Cheongju 361-763, Korea
| | | | | |
Collapse
|
82
|
miRNAs and Melanoma: How Are They Connected? Dermatol Res Pract 2011; 2012:528345. [PMID: 21860617 PMCID: PMC3154488 DOI: 10.1155/2012/528345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 06/15/2011] [Indexed: 12/15/2022] Open
Abstract
miRNAs are non-coding RNAs that bind to mRNA targets and disturb their stability and/or translation, thus acting in gene posttranscriptional regulation. It is predicted that over 30% of mRNAs are regulated by miRNAs. Therefore these molecules are considered essential in the processing of many biological responses, such as cell proliferation, apoptosis, and stress responsiveness. As miRNAs participate of virtually all cellular pathways, their deregulation is critical to cancer development. Consequently, loss or gain of miRNAs function may contribute to tumor progression. Little is known about the regulation of miRNAs and understanding the events that lead to changes in their expression may provide new perspectives for cancer treatment. Among distinct types of cancer, melanoma has special implications. It is characterized as a complex disease, originated from a malignant transformation of melanocytes. Despite being rare, its metastatic form is usually incurable, which makes melanoma the major death cause of all skin cancers. Some molecular pathways are frequently disrupted in melanoma, and miRNAs probably have a decisive role on these alterations. Therefore, this review aims to discuss new findings about miRNAs in melanoma fields, underlying epigenetic processes, and also to argue possibilities of using miRNAs in melanoma diagnosis and therapy.
Collapse
|
83
|
Abstract
Ion channels are involved in a variety of tumors. In particular, potassium channels are expressed abnormally in many cancer types, where their pharmacologic manipulation impairs tumor progression. Since this group of molecules has been successfully targeted for decades in other therapeutic areas, there is a significant body of knowledge on the pharmacology of potassium channels. Several groups of potassium channels with defined molecular identities have been proposed as candidates for therapeutic intervention. The strategies put forward range from classical small molecule blockade to gene therapy approaches, and include the use of potassium channels as targets for adjuvant therapy. We will discuss the reasons for these proposals and explore possible future developments.
Collapse
|
84
|
Kang YJ, Kim IY, Kim EH, Yoon MJ, Kim SU, Kwon TK, Choi KS. Paxilline enhances TRAIL-mediated apoptosis of glioma cells via modulation of c-FLIP, survivin and DR5. Exp Mol Med 2011; 43:24-34. [PMID: 21150246 DOI: 10.3858/emm.2011.43.1.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells while sparing normal cells. However, many cancer cells are resistant to TRAIL-induced cell death. Here, we report that paxilline, an indole alkaloid from Penicillium paxilli, can sensitize various glioma cells to TRAIL-mediated apoptosis. While treatment with TRAIL alone caused partial processing of caspase-3 to its p20 intermediate in TRAIL-resistant glioma cell lines, co-treatment with TRAIL and subtoxic doses of paxilline caused complete processing of caspase-3 into its active subunits. Paxilline treatment markedly upregulated DR5, a receptor of TRAIL, through a CHOP/GADD153-mediated process. In addition, paxilline treatment markedly downregulated the protein levels of the short form of the cellular FLICE-inhibitory protein (c-FLIPs) and the caspase inhibitor, survivin, through proteasome-mediated degradation. Taken together, these results show that paxilline effectively sensitizes glioma cells to TRAIL-mediated apoptosis by modulating multiple components of the death receptor-mediated apoptotic pathway. Interestingly, paxilline/TRAIL co-treatment did not induce apoptosis in normal astrocytes, nor did it affect the protein levels of CHOP, DR5 or survivin in these cells. Thus, combined treatment regimens involving paxilline and TRAIL may offer an attractive strategy for safely treating resistant gliomas.
Collapse
Affiliation(s)
- You Jung Kang
- Department of Molecular Science and Technology, Institute for Medical Sciences, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | |
Collapse
|
85
|
Desarnaud F, Geck P, Parkin C, Carpinito G, Makarovskiy AN. Gene expression profiling of the androgen independent prostate cancer cells demonstrates complex mechanisms mediating resistance to docetaxel. Cancer Biol Ther 2011; 11:204-12. [PMID: 21057205 DOI: 10.4161/cbt.11.2.13750] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanisms conferring resistance to docetaxel in prostate cancer patients remain partially understood. We generated docetaxel resistant derivatives of the androgen independent prostate cancer cell lines PC-3 and DU-145. Docetaxel rapidly induces DU-145 cell death via apoptosis and the drug resistant cells were produced by periodically exposing proliferating DU-145 cultures to small doses of docetaxel. In PC-3 cells docetaxel induces delayed cell death via mitotic catastrophe evident by profound multinucleation and formation of giant cells. Mononucleated progeny of the giant PC-3 cells shows significant resistance to docetaxel. Gene expression profiling of these docetaxel resistant PC-3 cells revealed sets of docetaxel inducible and constitutively expressed genes associated with major cancer pathways. A contradictory overlap with DU-145 docetaxel resistant cells was also found. Analyses suggested significant changes associated with apoptotic function, DNA repair, cell growth, survival and proliferation, metabolism, maintenance of cytoskeleton and extracellular matrix formation. These cellular processes often contribute to drug resistance and our study identified a set of genes managing this phenotype. Additional analyses of the drug resistant PC-3 cells using shRNA constructs determined direct relevance of Cyclin G2 to docetaxel resistance as well as prevention of multinucleation, whereas the knockdown of upregulated CYP1B1 showed no effect on either of these processes. Downregulated GBP1 was explored by ectopic overexpression and even though GBP1 has a potential to mediate resistance to docetaxel, it was not utilized in PC-3 cells. The results suggest complex combination of gene expression pattern changes that enables resistance to docetaxel while preventing death via multinucleation.
Collapse
Affiliation(s)
- Frank Desarnaud
- Department of Urology, Tufts Medical Center/Tufts University School of Medicine, Boston, MA, USA
| | | | | | | | | |
Collapse
|
86
|
Ruiz C, Holz DR, Oeggerli M, Schneider S, Gonzales IM, Kiefer JM, Zellweger T, Bachmann A, Koivisto PA, Helin HJ, Mousses S, Barrett MT, Azorsa DO, Bubendorf L. Amplification and overexpression of vinculin are associated with increased tumour cell proliferation and progression in advanced prostate cancer. J Pathol 2011; 223:543-52. [DOI: 10.1002/path.2828] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/21/2010] [Accepted: 11/23/2010] [Indexed: 02/01/2023]
|
87
|
Tajima N, Itokazu Y, Korpi ER, Somerharju P, Käkelä R. Activity of BK(Ca) channel is modulated by membrane cholesterol content and association with Na+/K+-ATPase in human melanoma IGR39 cells. J Biol Chem 2010; 286:5624-38. [PMID: 21135099 DOI: 10.1074/jbc.m110.149898] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interaction of large conductance Ca(2+)- and voltage-activated K(+) (BK(Ca)) channels with Na(+)/K(+)-ATPase, caveolin-1, and cholesterol was studied in human melanoma IGR39 cells. Functional BK(Ca) channels were enriched in caveolin-rich and detergent-resistant membranes, i.e. rafts, and blocking of the channels by a specific BK(Ca) blocker paxilline reduced proliferation of the cells. Disruption of rafts by selective depletion of cholesterol released BK(Ca) channels from these domains with a consequent increase in their activity. Consistently, cholesterol enrichment of the cells increased the proportion of BK(Ca) channels in rafts and decreased their activity. Immunocytochemical analysis showed that BK(Ca) channels co-localize with Na(+)/K(+)-ATPase in a cholesterol-dependent manner, thus suggesting their co-presence in rafts. Supporting this, ouabain, a specific blocker of Na(+)/K(+)-ATPase, inhibited BK(Ca) whole-cell current markedly in control cells but not in cholesterol-depleted ones. This inhibition required the presence of external Na(+). Collectively, these data indicate that the presence of Na(+)/K(+)-ATPase in rafts is essential for efficient functioning of BK(Ca) channels, presumably because the pump maintains a low intracellular Na(+) proximal to the BK(Ca) channel. In conclusion, cholesterol could play an important role in cellular ion homeostasis and thus modulate many cellular functions and cell proliferation.
Collapse
Affiliation(s)
- Nobuyoshi Tajima
- Department of Medical Biochemistry and Developmental Biology, Institute of Biomedicine, University of Helsinki, Helsinki FI-00014, Finland
| | | | | | | | | |
Collapse
|
88
|
Wu RS, Marx SO. The BK potassium channel in the vascular smooth muscle and kidney: α- and β-subunits. Kidney Int 2010; 78:963-74. [DOI: 10.1038/ki.2010.325] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
89
|
Mazar J, DeYoung K, Khaitan D, Meister E, Almodovar A, Goydos J, Ray A, Perera RJ. The regulation of miRNA-211 expression and its role in melanoma cell invasiveness. PLoS One 2010; 5:e13779. [PMID: 21072171 PMCID: PMC2967468 DOI: 10.1371/journal.pone.0013779] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 10/08/2010] [Indexed: 01/05/2023] Open
Abstract
The immediate molecular mechanisms behind invasive melanoma are poorly understood. Recent studies implicate microRNAs (miRNAs) as important agents in melanoma and other cancers. To investigate the role of miRNAs in melanoma, we subjected human melanoma cell lines to miRNA expression profiling, and report a range of variations in several miRNAs. Specifically, compared with expression levels in melanocytes, levels of miR-211 were consistently reduced in all eight non-pigmented melanoma cell lines we examined; they were also reduced in 21 out of 30 distinct melanoma samples from patients, classified as primary in situ, regional metastatic, distant metastatic, and nodal metastatic. The levels of several predicted target mRNAs of miR-211 were reduced in melanoma cell lines that ectopically expressed miR-211. In vivo target cleavage assays confirmed one such target mRNA encoded by KCNMA1. Mutating the miR-211 binding site seed sequences at the KCNMA1 3'-UTR abolished target cleavage. KCNMA1 mRNA and protein expression levels varied inversely with miR-211 levels. Two different melanoma cell lines ectopically expressing miR-211 exhibited significant growth inhibition and reduced invasiveness compared with the respective parental melanoma cell lines. An shRNA against KCNMA1 mRNA also demonstrated similar effects on melanoma cells. miR-211 is encoded within the sixth intron of TRPM1, a candidate suppressor of melanoma metastasis. The transcription factor MITF, important for melanocyte development and function, is needed for high TRPM1 expression. MITF is also needed for miR-211 expression, suggesting that the tumor-suppressor activities of MITF and/or TRPM1 may at least partially be due to miR-211's negative post transcriptional effects on the KCNMA1 transcript. Given previous reports of high KCNMA1 levels in metastasizing melanoma, prostate cancer and glioma, our findings that miR-211 is a direct posttranscriptional regulator of KCNMA1 expression as well as the dependence of this miRNA's expression on MITF activity, establishes miR-211 as an important regulatory agent in human melanoma.
Collapse
Affiliation(s)
- Joseph Mazar
- Sanford Burnham Medical Research Institute, Orlando, Florida, United States of America
| | - Katherine DeYoung
- Curtis and Elizabeth Anderson Cancer Institute, Savannah, Georgia, United States of America
| | - Divya Khaitan
- Sanford Burnham Medical Research Institute, Orlando, Florida, United States of America
| | - Edward Meister
- Curtis and Elizabeth Anderson Cancer Institute, Savannah, Georgia, United States of America
| | - Alvin Almodovar
- Sanford Burnham Medical Research Institute, Orlando, Florida, United States of America
| | - James Goydos
- Robert Wood Johnson Medical School, Cancer Institute of New Jersey, New Brunswick, New Jersey, United States of America
| | - Animesh Ray
- Keck Graduate Institute, Claremont, California, United States of America
| | - Ranjan J. Perera
- Sanford Burnham Medical Research Institute, Orlando, Florida, United States of America
- Curtis and Elizabeth Anderson Cancer Institute, Savannah, Georgia, United States of America
| |
Collapse
|
90
|
Jeffries O, Geiger N, Rowe ICM, Tian L, McClafferty H, Chen L, Bi D, Knaus HG, Ruth P, Shipston MJ. Palmitoylation of the S0-S1 linker regulates cell surface expression of voltage- and calcium-activated potassium (BK) channels. J Biol Chem 2010; 285:33307-33314. [PMID: 20693285 PMCID: PMC2963414 DOI: 10.1074/jbc.m110.153940] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/14/2010] [Indexed: 11/18/2022] Open
Abstract
S-palmitoylation is rapidly emerging as an important post-translational mechanism to regulate ion channels. We have previously demonstrated that large conductance calcium- and voltage-activated potassium (BK) channels are palmitoylated within an alternatively spliced (STREX) insert. However, these studies also revealed that additional site(s) for palmitoylation must exist outside of the STREX insert, although the identity or the functional significance of these palmitoylated cysteine residues are unknown. Here, we demonstrate that BK channels are palmitoylated at a cluster of evolutionary conserved cysteine residues (Cys-53, Cys-54, and Cys-56) within the intracellular linker between the S0 and S1 transmembrane domains. Mutation of Cys-53, Cys-54, and Cys-56 completely abolished palmitoylation of BK channels lacking the STREX insert (ZERO variant). Palmitoylation allows the S0-S1 linker to associate with the plasma membrane but has no effect on single channel conductance or the calcium/voltage sensitivity. Rather, S0-S1 linker palmitoylation is a critical determinant of cell surface expression of BK channels, as steady state surface expression levels are reduced by ∼55% in the C53:54:56A mutant. STREX variant channels that could not be palmitoylated in the S0-S1 linker also displayed significantly reduced cell surface expression even though STREX insert palmitoylation was unaffected. Thus our work reveals the functional independence of two distinct palmitoylation-dependent membrane interaction domains within the same channel protein and demonstrates the critical role of S0-S1 linker palmitoylation in the control of BK channel cell surface expression.
Collapse
Affiliation(s)
- Owen Jeffries
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Nina Geiger
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom; Pharmacology and Toxicology, Institute of Pharmacy, University Tuebingen, 72076 Tuebingen, Germany
| | - Iain C M Rowe
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Lijun Tian
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Heather McClafferty
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Lie Chen
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Danlei Bi
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Hans Guenther Knaus
- Division for Molecular and Cellular Pharmacology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University Innsbruck, Peter-Mayr Strasse 1, 6020 Innsbruck, Austria
| | - Peter Ruth
- Pharmacology and Toxicology, Institute of Pharmacy, University Tuebingen, 72076 Tuebingen, Germany
| | - Michael J Shipston
- From the Centre for Integrative Physiology, College of Medicine & Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom.
| |
Collapse
|
91
|
Ernest NJ, Logsdon NJ, McFerrin MB, Sontheimer H, Spiller SE. Biophysical properties of human medulloblastoma cells. J Membr Biol 2010; 237:59-69. [PMID: 20931182 DOI: 10.1007/s00232-010-9306-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Accepted: 09/22/2010] [Indexed: 01/01/2023]
Abstract
Medulloblastoma is a pediatric high-grade cerebellar malignancy derived from neuronal precursors. Although electrophysiologic characteristics of cerebellar granule neurons at all stages of cell development have been well described, such characterization has not been reported for medulloblastoma. In this study we attempt to characterize important electrophysiologic features of medulloblastoma that may distinguish it from the surrounding cerebellum. Using patient-derived cell lines and tumor tissues, we show that medulloblastoma cells have no inward Na+ current or transient K+ current involved in action potential generation and propagation, typically seen in granule neurons. Expression and function of calcium-activated, large-conductance K+ channels are diminished in medulloblastoma, judged by electrophysiology and Western analysis. The resting membrane potential of medulloblastoma cells in culture is quite depolarized compared to granule neurons. Interestingly, medulloblastoma cells express small, fast-inactivating calcium currents consistent with T-type calcium channels, but these channels are activated only from hyperpolarized potentials, which are unlikely to occur. Additionally, a background acid-sensitive K+ current is present with features characteristic of TASK1 or TASK3 channels, such as inhibition by ruthenium red. Western analysis confirms expression of TASK1 and TASK3. In describing the electrophysiologic characteristics of medulloblastoma, one can see features that resemble other high-grade malignancies as opposed to normal cerebellar granule neurons. This supports the notion that the malignant phenotype of medulloblastoma is characterized by unique changes in ion channel expression.
Collapse
Affiliation(s)
- Nola Jean Ernest
- Department of Pediatrics, University of Alabama School of Medicine, 1719 6th Ave. S., CIRC 252A, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
92
|
Savas S, Briollais L, Ibrahim-zada I, Jarjanazi H, Choi YH, Musquera M, Fleshner N, Venkateswaran V, Ozcelik H. A whole-genome SNP association study of NCI60 cell line panel indicates a role of Ca2+ signaling in selenium resistance. PLoS One 2010; 5:e12601. [PMID: 20830292 PMCID: PMC2935366 DOI: 10.1371/journal.pone.0012601] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 08/04/2010] [Indexed: 01/21/2023] Open
Abstract
Epidemiological studies have suggested an association between selenium intake and protection from a variety of cancer. Considering this clinical importance of selenium, we aimed to identify the genes associated with resistance to selenium treatment. We have applied a previous methodology developed by our group, which is based on the genetic and pharmacological data publicly available for the NCI60 cancer cell line panel. In short, we have categorized the NCI60 cell lines as selenium resistant and sensitive based on their growth inhibition (GI50) data. Then, we have utilized the Affymetrix 125K SNP chip data available and carried out a genome-wide case-control association study for the selenium sensitive and resistant NCI60 cell lines. Our results showed statistically significant association of four SNPs in 5q33–34, 10q11.2, 10q22.3 and 14q13.1 with selenium resistance. These SNPs were located in introns of the genes encoding for a kinase-scaffolding protein (AKAP6), a membrane protein (SGCD), a channel protein (KCNMA1), and a protein kinase (PRKG1). The knock-down of KCNMA1 by siRNA showed increased sensitivity to selenium in both LNCaP and PC3 cell lines. Furthermore, SNP-SNP interaction (epistasis) analysis indicated the interactions of the SNPs in AKAP6 with SGCD as well as SNPs in AKAP6 with KCNMA1 with each other, assuming additive genetic model. These genes were also all involved in the Ca2+ signaling, which has a direct role in induction of apoptosis and induction of apoptosis in tumor cells is consistent with the chemopreventive action of selenium. Once our findings are further validated, this knowledge can be translated into clinics where individuals who can benefit from the chemopreventive characteristics of the selenium supplementation will be easily identified using a simple DNA analysis.
Collapse
Affiliation(s)
- Sevtap Savas
- Fred A. Litwin Centre for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Laurent Briollais
- Prosserman Centre for Health Research, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Irada Ibrahim-zada
- Fred A. Litwin Centre for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hamdi Jarjanazi
- Fred A. Litwin Centre for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Yun Hee Choi
- Prosserman Centre for Health Research, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mireia Musquera
- Division of Urology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Neil Fleshner
- Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Vasundara Venkateswaran
- Division of Urology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- * E-mail: (VV); (HO)
| | - Hilmi Ozcelik
- Fred A. Litwin Centre for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (VV); (HO)
| |
Collapse
|
93
|
Calcium-activated potassium channels BK and IK1 are functionally expressed in human gliomas but do not regulate cell proliferation. PLoS One 2010; 5:e12304. [PMID: 20808839 PMCID: PMC2924897 DOI: 10.1371/journal.pone.0012304] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Accepted: 07/25/2010] [Indexed: 01/15/2023] Open
Abstract
Gliomas are morbid brain tumors that are extremely resistant to available chemotherapy and radiology treatments. Some studies have suggested that calcium-activated potassium channels contribute to the high proliferative potential of tumor cells, including gliomas. However, other publications demonstrated no role for these channels or even assigned them antitumorogenic properties. In this work we characterized the expression and functional contribution to proliferation of Ca2+-activated K+ channels in human glioblastoma cells. Quantitative RT-PCR detected transcripts for the big conductance (BK), intermediate conductance (IK1), and small conductance (SK2) K+ channels in two glioblastoma-derived cell lines and a surgical sample of glioblastoma multiforme. Functional expression of BK and IK1 in U251 and U87 glioma cell lines and primary glioma cultures was verified using whole-cell electrophysiological recordings. Inhibitors of BK (paxilline and penitrem A) and IK1 channels (clotrimazole and TRAM-34) reduced U251 and U87 proliferation in an additive fashion, while the selective blocker of SK channels UCL1848 had no effect. However, the antiproliferative properties of BK and IK1 inhibitors were seen at concentrations that were higher than those necessary to inhibit channel activity. To verify specificity of pharmacological agents, we downregulated BK and IK1 channels in U251 cells using gene-specific siRNAs. Although siRNA knockdowns caused strong reductions in the BK and IK1 current densities, neither single nor double gene silencing significantly affected rates of proliferation. Taken together, these results suggest that Ca2+-activated K+ channels do not play a critical role in proliferation of glioma cells and that the effects of pharmacological inhibitors occur through their off-target actions.
Collapse
|
94
|
LRRC26 auxiliary protein allows BK channel activation at resting voltage without calcium. Nature 2010; 466:513-6. [PMID: 20613726 DOI: 10.1038/nature09162] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 05/07/2010] [Indexed: 11/08/2022]
Abstract
Large-conductance, voltage- and calcium-activated potassium (BK, or K(Ca)1.1) channels are ubiquitously expressed in electrically excitable and non-excitable cells, either as alpha-subunit (BKalpha) tetramers or together with tissue specific auxiliary beta-subunits (beta1-beta4). Activation of BK channels typically requires coincident membrane depolarization and elevation in free cytosolic Ca(2+) concentration ([Ca(2+)](i)), which are not physiological conditions for most non-excitable cells. Here we present evidence that in non-excitable LNCaP prostate cancer cells, BK channels can be activated at negative voltages without rises in [Ca(2+)](i) through their complex with an auxiliary protein, leucine-rich repeat (LRR)-containing protein 26 (LRRC26). LRRC26 modulates the gating of a BK channel by enhancing the allosteric coupling between voltage-sensor activation and the channel's closed-open transition. This finding reveals a novel auxiliary protein of a voltage-gated ion channel that gives an unprecedentedly large negative shift ( approximately -140 mV) in voltage dependence and provides a molecular basis for activation of BK channels at physiological voltages and calcium levels in non-excitable cells.
Collapse
|
95
|
Park JH, Park SJ, Chung MK, Jung KH, Choi MR, Kim Y, Chai YG, Kim SJ, Park KS. High expression of large-conductance Ca2+-activated K+ channel in the CD133+ subpopulation of SH-SY5Y neuroblastoma cells. Biochem Biophys Res Commun 2010; 396:637-42. [PMID: 20438714 DOI: 10.1016/j.bbrc.2010.04.142] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 04/27/2010] [Indexed: 01/27/2023]
Abstract
Solid tumors contain a population of cancer stem cells (CSCs), and CD133 is widely used as a CSCs marker. We investigated the differences between CD133(+) and CD133(-) cells from the neuroblastoma cell line SH-SY5Y in terms of the expressions of voltage-gated ion channels. A CD133(+) enriched (>60%) population was isolated, and a subsequent whole-cell voltage-clamp study showed that these cells predominantly express TEA-sensitive outward K(+) currents (I(K,TEA)) and TTX-sensitive voltage-gated inward Na(+) currents (I(Na)). Cell-attached single channel recordings demonstrated higher density of large-conductance (155pS) channel in CD133(+) cells than in CD133(-) cells. The TEA-sensitivity and single channel conductance indicated the large-conductance Ca(2+)-activated K(+) channels (BK(Ca)). Furthermore, RT-PCR analysis of 22 transcripts of voltage-gated ion channels in SH-SY5Y cells showed the expressions of Cav1.3, Kir2.1, Kv1.4, Kv2.1, Kv4.2, Kv7.1, BK(Ca), and Nav1.7, and those of BK(Ca) and Nav1.7 were higher in CD133(+) than in CD133(-) cells. In addition, the increase of cytosolic Ca(2+) concentration ([Ca(2+)](c)) in response to ionomycin (a Ca(2+) ionophore) was higher and more sustained in CD133(+) than in CD133(-) cells. Plausibly membrane hyperpolarization via BK(Ca) might be responsible for the augmented Ca(2+) influx observed in CD133(+) cells. The physiological implications of the differential expression of BK(Ca) and Nav1.7 in CSCs require further investigation.
Collapse
Affiliation(s)
- Ji Hyun Park
- Institute Division of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Gordon E, Semus SF, Lozinskaya IM, Lin Z, Xu X. Characterizing the Role of Thr352 in the Inhibition of the Large Conductance Ca2+-Activated K+ Channels by 1-[1-Hexyl-6-(methyloxy)-1H-indazol-3-yl]-2-methyl-1-propanone. J Pharmacol Exp Ther 2010; 334:402-9. [DOI: 10.1124/jpet.110.166017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
97
|
Humbert L, Chevrette M. Somatic Molecular Genetics of Prostate Cancer. MALE REPRODUCTIVE CANCERS 2010:143-180. [DOI: 10.1007/978-1-4419-0449-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
98
|
Intermediate-conductance Ca2+-activated K+ channels (IKCa1) regulate human prostate cancer cell proliferation through a close control of calcium entry. Oncogene 2009; 28:1792-806. [DOI: 10.1038/onc.2009.25] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
99
|
Ohya S, Kimura K, Niwa S, Ohno A, Kojima Y, Sasaki S, Kohri K, Imaizumi Y. Malignancy Grade–Dependent Expression of K+-Channel Subtypes in Human Prostate Cancer. J Pharmacol Sci 2009; 109:148-51. [DOI: 10.1254/jphs.08208sc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
100
|
Sirintrapun SJ, Parwani AV. Molecular Pathology of the Genitourinary Tract: Prostate and Bladder. Surg Pathol Clin 2008; 1:211-36. [PMID: 26837907 DOI: 10.1016/j.path.2008.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The knowledge of cellular mechanisms in tumors of the prostate and bladder has grown exponentially. Molecular technologies have led to the discovery of TMPRSS2 in prostate cancer and the molecular pathways distinguishing low- and high-grade urothelial neoplasms. UroVysion with fluorescence in situ hybridization is already commonplace as an adjunct to cytologic diagnosis of urothelial neoplasms. This trend portends the future in which classification and diagnosis of tumors of the prostate and bladder through morphologic analysis will be supplemented by molecular information correlating with prognosis and targeted therapy. This article outlines tumor molecular pathology of the prostate and bladder encompassing current genomic, epigenomic, and proteonomic findings.
Collapse
Affiliation(s)
- S Joseph Sirintrapun
- Pathology Informatics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh Medical Center Shadyside Hospital, Room WG 07, 5230 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|