51
|
Zeng W, Liu Q, Chen Z, Wu X, Zhong Y, Wu J. Silencing of hERG1 Gene Inhibits Proliferation and Invasion, and Induces Apoptosis in Human Osteosarcoma Cells by Targeting the NF-κB Pathway. J Cancer 2016; 7:746-57. [PMID: 27076857 PMCID: PMC4829562 DOI: 10.7150/jca.13289] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 02/11/2016] [Indexed: 02/06/2023] Open
Abstract
Recently, the human ether à go-go (eag) related gene 1 (hERG1) channel, a member of the voltage-dependent potassium channel (Kv) family, was determined to have a critical role in cancer cell proliferation, invasion, tumorigenesis and apoptosis. However, the expression levels and functions of hERG1 in osteosarcoma cells remain poorly characterized. In this study, hERG1 transcript and protein levels in osteosarcoma cells and tissues were measured using semi-quantitative real time PCR (RT-PCR), Western blot, and immunohistochemistry. The effects of hERG1 knockdown on osteosarcoma cell proliferation, apoptosis and invasion were examined using CCK-8, colony formation, flow cytometry, caspase-3 activity, wound healing and transwell based assays. Furthermore, semi-quantitative RT-PCR, Western blot and a luciferase reporter assay were used to assess the effects of hERG1 inhibition on the nuclear factor-κB (NF-κB) pathway. In addition, the effect of NF-κB p65-siRNA and NF-κB p65 expression on the survival of osteosarcoma cells was investigated. Through this work, a relationship for hERG1 with the NF-κB pathway was identified. Osteosarcoma cells and tissues were found to express high levels of hERG1. Knockdown of hERG1 significantly suppressed cellular proliferation and invasion, and induced apoptosis, while inhibition of hERG1 significantly decreased activation of NF-κB. Overall, hERG1 may stimulate nuclear translocation of p65, thus regulating the NF-κB pathway through the activation of the hERG1/beta1 integrin complex and PI3K/AKT signaling. Taken together, these results demonstrate that hERG1 is necessary for regulation of osteosarcoma cellular proliferation, apoptosis and migration. Furthermore, this regulation by hERG1 is, at least in part, through mediation of the NF-κB pathway.
Collapse
Affiliation(s)
- Wenrong Zeng
- 1. Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, People's Republic of China
| | - Qingjun Liu
- 1. Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, People's Republic of China
| | - Zhida Chen
- 1. Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, People's Republic of China
| | - Xinyu Wu
- 2. Department of Neurology, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, 363000, People's Republic of China
| | - Yuanfu Zhong
- 3. Department of central laboratory, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, 363000, People's Republic of China
| | - Jin Wu
- 1. Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, People's Republic of China
| |
Collapse
|
52
|
Crottès D, Rapetti-Mauss R, Alcaraz-Perez F, Tichet M, Gariano G, Martial S, Guizouarn H, Pellissier B, Loubat A, Popa A, Paquet A, Presta M, Tartare-Deckert S, Cayuela ML, Martin P, Borgese F, Soriani O. SIGMAR1 Regulates Membrane Electrical Activity in Response to Extracellular Matrix Stimulation to Drive Cancer Cell Invasiveness. Cancer Res 2016; 76:607-18. [PMID: 26645564 DOI: 10.1158/0008-5472.can-15-1465] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/27/2015] [Indexed: 01/12/2023]
Abstract
The sigma 1 receptor (Sig1R) is a stress-activated chaperone that regulates ion channels and is associated with pathologic conditions, such as stroke, neurodegenerative diseases, and addiction. Aberrant expression levels of ion channels and Sig1R have been detected in tumors and cancer cells, such as myeloid leukemia and colorectal cancer, but the link between ion channel regulation and Sig1R overexpression during malignancy has not been established. In this study, we found that Sig1R dynamically controls the membrane expression of the human voltage-dependent K(+) channel human ether-à-go-go-related gene (hERG) in myeloid leukemia and colorectal cancer cell lines. Sig1R promoted the formation of hERG/β1-integrin signaling complexes upon extracellular matrix stimulation, triggering the activation of the PI3K/AKT pathway. Consequently, the presence of Sig1R in cancer cells increased motility and VEGF secretion. In vivo, Sig1R expression enhanced the aggressiveness of tumor cells by potentiating invasion and angiogenesis, leading to poor survival. Collectively, our findings highlight a novel function for Sig1R in mediating cross-talk between cancer cells and their microenvironment, thus driving oncogenesis by shaping cellular electrical activity in response to extracellular signals. Given the involvement of ion channels in promoting several hallmarks of cancer, our study also offers a potential strategy to therapeutically target ion channel function through Sig1R inhibition.
Collapse
Affiliation(s)
- David Crottès
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France. Department of Physiology, University of California, San Francisco, San Francisco, California
| | - Raphael Rapetti-Mauss
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Francisca Alcaraz-Perez
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Mélanie Tichet
- Université Nice Sophia Antipolis, C3M, Inserm U1065, Nice, France
| | - Giuseppina Gariano
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sonia Martial
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Hélène Guizouarn
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Bernard Pellissier
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Agnès Loubat
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Alexandra Popa
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Agnès Paquet
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Marco Presta
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Maria Luisa Cayuela
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Patrick Martin
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Franck Borgese
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Olivier Soriani
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France.
| |
Collapse
|
53
|
Menéndez ST, Villaronga MÁ, Rodrigo JP, Álvarez-Teijeiro S, Urdinguio RG, Fraga MF, Suárez C, García-Pedrero JM. HERG1A potassium channel is the predominant isoform in head and neck squamous cell carcinomas: evidence for regulation by epigenetic mechanisms. Sci Rep 2016; 6:19666. [PMID: 26785772 PMCID: PMC4726400 DOI: 10.1038/srep19666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/16/2015] [Indexed: 11/09/2022] Open
Abstract
Evidences indicate that HERG1 voltage-gated potassium channel is frequently aberrantly expressed in various cancers including head and neck squamous cell carcinomas (HNSCC), representing a clinically and biologically relevant feature during disease progression and a potential therapeutic target. The present study further and significantly extends these data investigating for the first time the expression and individual contribution of HERG1 isoforms, their clinical significance during disease progression and also the underlying regulatory mechanisms. Analysis of HERG1A and HERG1B expression using real-time RT-PCR consistently showed that HERG1A is the predominant isoform in ten HNSCC-derived cell lines tested. HERG2 and HERG3 were also detected. Immunohistochemical analysis of HERG1A expression on 133 HNSCC specimens demonstrated that HERG1A expression increased during tumour progression and correlated significantly with reduced disease-specific survival. Furthermore, our study provides original evidence supporting the involvement of histone acetylation (i.e. H3Ac and H4K16Ac activating marks) in the regulation of HERG1 expression in HNSCC. Interestingly, this mechanism was also found to regulate the expression of another oncogenic channel (Kv3.4) as well as HERG2 and HERG3. These data demonstrate that HERG1A is the predominant and disease-relevant isoform in HNSCC progression, while histone acetylation emerges as an important regulatory mechanism underlying Kv gene expression.
Collapse
Affiliation(s)
- Sofía T Menéndez
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - M Ángeles Villaronga
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Juan P Rodrigo
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Saúl Álvarez-Teijeiro
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Rocío G Urdinguio
- Unidad de Epigenética del Cáncer, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Mario F Fraga
- Unidad de Epigenética del Cáncer, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain.,Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| | - Carlos Suárez
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Juana M García-Pedrero
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
54
|
Simon OJ, Müntefering T, Grauer OM, Meuth SG. The role of ion channels in malignant brain tumors. J Neurooncol 2015; 125:225-35. [PMID: 26334315 DOI: 10.1007/s11060-015-1896-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 08/14/2015] [Indexed: 12/15/2022]
Abstract
Malignant gliomas are the most common primary brain tumors and have poor clinical prognosis, despite multimodal therapeutic strategies. In recent years, ion channels have emerged as major players in tumor pathophysiology regarding all hallmarks of cancer. Since ion channels are easily accessible structures, they may prove to be effective targets for canner therapy, although their broad expression pattern and role in physiological processes should be taken into consideration. This review summarizes the current knowledge on the role of ion channels in the pathophysiology of malignant gliomas, especially glioblastoma, and evaluates their potential role in targeted antiglioma therapy.
Collapse
Affiliation(s)
- Ole J Simon
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| | - Thomas Müntefering
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| |
Collapse
|
55
|
Lastraioli E, Lottini T, Bencini L, Bernini M, Arcangeli A. hERG1 Potassium Channels: Novel Biomarkers in Human Solid Cancers. BIOMED RESEARCH INTERNATIONAL 2015; 2015:896432. [PMID: 26339650 PMCID: PMC4538961 DOI: 10.1155/2015/896432] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/16/2015] [Accepted: 02/24/2015] [Indexed: 01/05/2023]
Abstract
Because of their high incidence and mortality solid cancers are a major health problem worldwide. Although several new biomarkers and potential targets for therapy have been identified through biomolecular research in the last years, the effects on patients' outcome are still unsatisfactory. Increasing evidence indicates that hERG1 potassium channels are overexpressed in human primary cancers of different origin and several associations between hERG1 expression and clinicopathological features and/or outcome are emerging. Aberrant hERG1 expression may be exploited either for early diagnosis (especially in those cancers where it is expressed in the initial steps of tumor progression) or for therapy purposes. Indeed, hERG1 blockage impairs tumor cell growth both in vitro and in vivo in preclinical mouse model. hERG1-based tumor therapy in humans, however, encounters the major hindrance of the potential cardiotoxicity that many hERG1 blockers exert. In this review we focus on recent advances in translational research in some of the most frequent human solid cancers (breast, endometrium, ovary, pancreas, esophagus, stomach, and colorectum) that have been shown to express hERG1 and that are a major health problem.
Collapse
Affiliation(s)
- Elena Lastraioli
- Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Tiziano Lottini
- Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Lapo Bencini
- General Surgery and Surgical Oncology, Careggi University Hospital, Largo A Brambilla 3, 50134 Florence, Italy
| | - Marco Bernini
- Breast Unit Surgery, Department of Oncology, Careggi University Hospital, Largo A Brambilla 3, 50134 Florence, Italy
| | - Annarosa Arcangeli
- Experimental and Clinical Medicine, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
56
|
Arcangeli A, Becchetti A. Novel perspectives in cancer therapy: Targeting ion channels. Drug Resist Updat 2015; 21-22:11-9. [DOI: 10.1016/j.drup.2015.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 06/24/2015] [Accepted: 06/27/2015] [Indexed: 01/04/2023]
|
57
|
Effect of celastrol on growth inhibition of prostate cancer cells through the regulation of hERG channel in vitro. BIOMED RESEARCH INTERNATIONAL 2015; 2015:308475. [PMID: 25866772 PMCID: PMC4383143 DOI: 10.1155/2015/308475] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 07/28/2014] [Indexed: 11/17/2022]
Abstract
Objective. To explore the antiprostate cancer effects of Celastrol on prostate cancer cells' proliferation, apoptosis, and cell cycle distribution, as well as the correlation to the regulation of hERG. Methods. DU145 cells were treated with various concentrations of Celastrol (0.25–16.0 μmol/L) for 0–72 hours. MTT assay was used to evaluate the inhibition effect of Celastrol on the growth of DU145 cells. Cell apoptosis was detected through both Annexin-V FITC/PI double-labeled cytometry and Hoechst 33258. Cell cycle regulation was examined by a propidium iodide method. Western blot and RT-PCR technologies were applied to assess the expression level of hERG in DU145 cells. Results. Celastrol presented striking growth inhibition and apoptosis induction potency on DU145 cells in vitro in a time- and dose-dependent manner. The IC50 value of Celastrol for 24 hours was 2.349 ± 0.213 μmol/L. Moreover, Celastrol induced DU145 cell apoptosis in a cell cycle-dependent manner, which means Celastrol could arrest DU145 cells in G0/G1 phase; accordingly, cells in S phase decreased gradually and no obvious changes were found in G2/M phase cells. Through transmission electron microscope, apoptotic bodies containing nuclear fragments were found in Celastrol-treated DU145 cells. Overexpression of hERG channel was found in DU145 cells, while Celastrol could downregulate it at both protein and mRNA level in a dose-dependent manner (P < 0.01). Conclusions. Celastrol exhibits its antiprostate cancer effects partially through the downregulation of the expression level of hERG channel in DU145 cells, suggesting that Celastrol may be a potential agent against prostate cancer with a mechanism of blocking the hERG channel.
Collapse
|
58
|
Gasparoli L, D'Amico M, Masselli M, Pillozzi S, Caves R, Khuwaileh R, Tiedke W, Mugridge K, Pratesi A, Mitcheson JS, Basso G, Becchetti A, Arcangeli A. New pyrimido-indole compound CD-160130 preferentially inhibits the KV11.1B isoform and produces antileukemic effects without cardiotoxicity. Mol Pharmacol 2015; 87:183-96. [PMID: 25411366 DOI: 10.1124/mol.114.094920] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
KV11.1 (hERG1) channels are often overexpressed in human cancers. In leukemias, KV11.1 regulates pro-survival signals that promote resistance to chemotherapy, raising the possibility that inhibitors of KV11.1 could be therapeutically beneficial. However, because of the role of KV11.1 in cardiac repolarization, blocking these channels may cause cardiac arrhythmias. We show that CD-160130, a novel pyrimido-indole compound, blocks KV11.1 channels with a higher efficacy for the KV11.1 isoform B, in which the IC50 (1.8 μM) was approximately 10-fold lower than observed in KV11.1 isoform A. At this concentration, CD-160130 also had minor effects on Kir2.1, KV 1.3, Kv1.5, and KCa3.1. In vitro, CD-160130 induced leukemia cell apoptosis, and could overcome bone marrow mesenchymal stromal cell (MSC)-induced chemoresistance. This effect was caused by interference with the survival signaling pathways triggered by MSCs. In vivo, CD-160130 produced an antileukemic activity, stronger than that caused by cytarabine. Consistent with its atypical target specificity, CD-160130 did not bind to the main binding site of the arrhythmogenic KV11.1 blockers (the Phe656 pore residue). Importantly, in guinea pigs CD-160130 produced neither alteration of the cardiac action potential shape in dissociated cardiomyocytes nor any lengthening of the QT interval in vivo. Moreover, CD-160130 had no myelotoxicity on human bone marrow-derived cells. Therefore, CD-160130 is a promising first-in-class compound to attempt oncologic therapy without cardiotoxicity, based on targeting KV11.1. Because leukemia and cardiac cells tend to express different ratios of the A and B KV11.1 isoforms, the pharmacological properties of CD-160130 may depend, at least in part, on isoform specificity.
Collapse
Affiliation(s)
- Luca Gasparoli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Massimo D'Amico
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Marika Masselli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Rachel Caves
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Rawan Khuwaileh
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Wolfgang Tiedke
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Kenneth Mugridge
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Alessandro Pratesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - John S Mitcheson
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Giuseppe Basso
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Andrea Becchetti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (L.G., S.P., A.A.); Department of Chemistry "Ugo Schiff," University of Florence, Florence, Italy (M.M., A.P.); DI.V.A.L. Toscana srl, Sesto Fiorentino, Italy (M.D.A., M.M.); Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom (R.C., R.K., J.S.M.); BlackSwan Pharma GmbH, Leipzig, Germany (W.T., K.M.); Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy (G.B.); and Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy (A.B.)
| |
Collapse
|
59
|
Involvement of potassium channels in the progression of cancer to a more malignant phenotype. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2477-92. [PMID: 25517985 DOI: 10.1016/j.bbamem.2014.12.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 12/01/2014] [Accepted: 12/08/2014] [Indexed: 12/22/2022]
Abstract
Potassium channels are a diverse group of pore-forming transmembrane proteins that selectively facilitate potassium flow through an electrochemical gradient. They participate in the control of the membrane potential and cell excitability in addition to different cell functions such as cell volume regulation, proliferation, cell migration, angiogenesis as well as apoptosis. Because these physiological processes are essential for the correct cell function, K+ channels have been associated with a growing number of diseases including cancer. In fact, different K+ channel families such as the voltage-gated K+ channels, the ether à-go-go K+ channels, the two pore domain K+ channels and the Ca2+-activated K+ channels have been associated to tumor biology. Potassium channels have a role in neoplastic cell-cycle progression and their expression has been found abnormal in many types of tumors and cancer cells. In addition, the expression and activity of specific K+ channels have shown a significant correlation with the tumor malignancy grade. The aim of this overview is to summarize published data on K+ channels that exhibit oncogenic properties and have been linked to a more malignant cancer phenotype. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
|
60
|
Wang J, Li Y, Jiang C. MiR-133b contributes to arsenic-induced apoptosis in U251 glioma cells by targeting the hERG channel. J Mol Neurosci 2014; 55:985-94. [PMID: 25355491 DOI: 10.1007/s12031-014-0455-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/21/2014] [Indexed: 12/19/2022]
Abstract
Substantial evidence indicates that the human ether-a-go-go-related gene potassium channel (hERG, Kv11.1, KCNH2) is overexpressed in human glioblastoma multiforme (GBM) specimens and plays an essential role in the malignant proliferation of glioma cells. However, its upstream regulator in glioma cells is not fully elucidated. The present study was designed to determine whether the expression of hERG gene is regulated by miR-133b or miR-34a, thereby contributing to the anti-proliferation effect of arsenic trioxide (ATO) in U251 human glioma cells. Real-time polymerase chain reactions (qRT-PCR) and Western blot results demonstrated that hERG mRNA and protein levels were dramatically upregulated in clinical GBM specimens. Conversely, both miR-133b and miR-34a were markedly downregulated in clinical GBM specimens by qRT-PCR. The hERG gene was a direct target of miR-133b and miR-34a by bioinformatics analyses and luciferase reporter assays. Moreover, ATO, which is an emerging chemotherapy drug for glioma disease, remarkably elevated the level of miR-133b, but not miR-34a in U251 glioma cells. The level of miR-133b upstream transactivator serum response factor (SRF) was also suppressed by ATO. The transfection of anti-miR-133b oligonucleotide (AMO-133b) remarkably prevented the decrease of hERG protein by 5 μM ATO treatment for 24 h in U251 cells, whereas anti-miR-34a oligonucleotide (AMO-34a) did not exhibit recuperated effect. Finally, the transient overexpression by miR-133b mimics and treatment with the hERG channel-specific blocker E4031 markedly facilitated the ATO inhibition of proliferation of and induced apoptosis in U251 cells, whereas AMO-miR-133b attenuated these changes. Our study provided the evidence for the pathological role of miR-133b and miR-34a in the development of GBM and thus expanded our understanding of the hERG gene expression and ATO chemotherapeutic roles of miRNAs. Targeting miR-133b/hERG pathway may be a new strategy for chemotherapy of malignant gliomas.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, People's Republic of China
| | | | | |
Collapse
|
61
|
Mitcheson J, Arcangeli A. The Therapeutic Potential of hERG1 K+ Channels for Treating Cancer and Cardiac Arrhythmias. ION CHANNEL DRUG DISCOVERY 2014. [DOI: 10.1039/9781849735087-00258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
hERG potassium channels present pharmacologists and medicinal chemists with a dilemma. On the one hand hERG is a major reason for drugs being withdrawn from the market because of drug induced long QT syndrome and the associated risk of inducing sudden cardiac death, and yet hERG blockers are still widely used in the clinic to treat cardiac arrhythmias. Moreover, in the last decade overwhelming evidence has been provided that hERG channels are aberrantly expressed in cancer cells and that they contribute to tumour cell proliferation, resistance to apoptosis, and neoangiogenesis. Here we provide an overview of the properties of hERG channels and their role in excitable cells of the heart and nervous system as well as in cancer. We consider the therapeutic potential of hERG, not only with regard to the negative impact due to drug induced long QT syndrome, but also its future potential as a treatment in the fight against cancer.
Collapse
Affiliation(s)
- John Mitcheson
- University of Leicester, Department of Cell Physiology and Pharmacology, Medical Sciences Building University Road Leicester LE1 9HN UK
| | - Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Florence Viale GB Morgagni, 50 50134 Firenze Italy
| |
Collapse
|
62
|
Pier DM, Shehatou GSG, Giblett S, Pullar CE, Trezise DJ, Pritchard CA, Challiss RAJ, Mitcheson JS. Long-term channel block is required to inhibit cellular transformation by human ether-à-go-go-related gene (hERG1) potassium channels. Mol Pharmacol 2014; 86:211-21. [PMID: 24830940 PMCID: PMC4127929 DOI: 10.1124/mol.113.091439] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 05/14/2014] [Indexed: 11/22/2022] Open
Abstract
Both human ether-à-go-go-related gene (hERG1) and the closely related human ether-à-go-go (hEAG1) channel are aberrantly expressed in a large proportion of human cancers. In the present study, we demonstrate that transfection of hERG1 into mouse fibroblasts is sufficient to induce many features characteristic of malignant transformation. An important finding of this work is that this transformation could be reversed by chronic incubation (for 2-3 weeks) with the hERG channel blocker dofetilide (100 nM), whereas more acute applications (for 1-2 days) were ineffective. The hERG1 expression resulted in a profound loss of cell contact inhibition, multiple layers of overgrowing cells, and high saturation densities. Cells also changed from fibroblast-like to a more spindle-shaped morphology, which was associated with a smaller cell size, a dramatic increase in cell polarization, a reduction in the number of actin stress fibers, and less punctate labeling of focal adhesions. Analysis of single-cell migration and scratch-wound closure clearly demonstrated that hERG1-expressing cells migrated more rapidly than vector-transfected control cells. In contrast to previous studies on hEAG1, there were no increases in rates of proliferation, or loss of growth factor dependency; however, hERG1-expressing cells were capable of substrate-independent growth. Allogeneic transplantation of hERG1-expressing cells into nude mice resulted in an increased incidence of tumors. In contrast to hEAG1, the mechanism of cellular transformation is dependent on ion conduction. Trafficking-deficient and conduction-deficient hERG1 mutants also prevented cellular transformation. These results provide evidence that hERG1 expression is sufficient to induce cellular transformation by a mechanism distinct from hEAG1. The most important conclusion of this study is that selective hERG1 channel blockers have therapeutic potential in the treatment of hERG1-expressing cancers.
Collapse
Affiliation(s)
- David M Pier
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - George S G Shehatou
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Susan Giblett
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Christine E Pullar
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Derek J Trezise
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Catrin A Pritchard
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - R A John Challiss
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - John S Mitcheson
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| |
Collapse
|
63
|
Wei X, Sun H, Yan H, Zhang C, Zhang S, Liu X, Hua N, Ma X, Zheng J. ZC88, a novel 4-amino piperidine analog, inhibits the growth of neuroblastoma cells through blocking hERG potassium channel. Cancer Biol Ther 2014; 14:450-7. [PMID: 23917377 DOI: 10.4161/cbt.24423] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Many studies have provided convincing evidence for hERG as an important diagnostic and prognostic factor in human cancers, as well as a useful target for antineoplastic therapy. Our previous study also revealed that knockdown of herg gene expression by shRNA interference inhibited the growth of neuroblastoma cells in vitro and in vivo. In the experiment, a novel 4-amino piperidine analog, ZC88, was examined for its effect on hERG potassium channels and its antitumor potency was observed in vitro and in vivo. The results showed that ZC88 could block hERG1 and hERG1b channels expressed in Xenopus oocytes in a concentration-dependent manner. ZC88 displayed significant antiproliferative activity in several tumor cell lines and the tumor cells with higher expression of hERG presented higher sensitivity to ZC88. The mitotic progression of tumor cells was markedly suppressed in the presence of ZC88 through arresting cells in G₀/G₁ phase. ZC88 significantly inhibited the tumor growth in nude mice at a dosage with slight influence on the cardiac QT interval. The antitumor effect of ZC88 was correlated at least partly with its blockage of hERG channels, which implicated a positive role of hERG potassium channel in tumor cell proliferation.
Collapse
Affiliation(s)
- Xiaoli Wei
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Crociani O, Lastraioli E, Boni L, Pillozzi S, Romoli MR, D'Amico M, Stefanini M, Crescioli S, Masi A, Taddei A, Bencini L, Bernini M, Farsi M, Beghelli S, Scarpa A, Messerini L, Tomezzoli A, Vindigni C, Morgagni P, Saragoni L, Giommoni E, Gasperoni S, Di Costanzo F, Roviello F, De Manzoni G, Bechi P, Arcangeli A. hERG1 channels regulate VEGF-A secretion in human gastric cancer: clinicopathological correlations and therapeutical implications. Clin Cancer Res 2014; 20:1502-1512. [PMID: 24449824 DOI: 10.1158/1078-0432.ccr-13-2633] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE hERG1 channels are aberrantly expressed in several types of human cancers, where they affect different aspects of cancer cell behavior. A thorough analysis of the functional role and clinical significance of hERG1 channels in gastric cancer is still lacking. EXPERIMENTAL DESIGN hERG1 expression was tested in a wide (508 samples) Italian cohort of surgically resected patients with gastric cancer, by immunohistochemistry and real-time quantitative PCR. The functional link between hERG1 and the VEGF-A was studied in different gastric cancer cell lines. The effects of hERG1 and VEGF-A inhibition were evaluated in vivo in xenograft mouse models. RESULTS hERG1 was positive in 69% of the patients and positivity correlated with Lauren's intestinal type, fundus localization of the tumor, G1-G2 grading, I and II tumor-node-metastasis stage, and VEGF-A expression. hERG1 activity modulated VEGF-A secretion, through an AKT-dependent regulation of the transcriptional activity of the hypoxia inducible factor. Treatment of immunodeficient mice xenografted with human gastric cancer cells, with a combination of hERG1 blockers and anti-VEGF-A antibodies, impaired tumor growth more than single-drug treatments. CONCLUSION Our results show that hERG1 (i) is aberrantly expressed in human gastric cancer since its early stages; (ii) drives an intracellular pathway leading to VEGF-A secretion; (iii) can be exploited to identify a gastric cancer patients' group where a combined treatment with antiangiogenic drugs and noncardiotoxic hERG1 inhibitors could be proposed.
Collapse
Affiliation(s)
- Olivia Crociani
- Authors' Affiliations: Department of Clinical and Experimental Medicine; Surgery and Translational Medicine, University of Florence; Clinical Trials Coordinating Center; General Surgery and Surgical Oncology; Medical Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence; Department of Pathology and Diagnostics, Division of Surgery, University of Verona; Pathology Division, Borgo Trento Hospital, Verona; Pathology Division, Azienda Ospedaliero-Universitaria Senese, Department of General Surgery and Oncology, University of Siena, Siena; and General Surgery and Division of Pathology, Morgagni-Pierantoni Hospital, Forlì, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Pakladok T, Hosseinzadeh Z, Almilaji A, Lebedeva A, Shumilina E, Alesutan I, Lang F. Up-regulation of hERG K⁺ channels by B-RAF. PLoS One 2014; 9:e87457. [PMID: 24475291 PMCID: PMC3903650 DOI: 10.1371/journal.pone.0087457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/26/2013] [Indexed: 11/19/2022] Open
Abstract
Human ether-a-go-go related-gene K⁺ channels (hERG) participate in the regulation of tumor cell proliferation and apoptosis. HERG channel activity is up-regulated by growth factors. Kinases sensitive to growth factor signaling include the serine/threonine protein kinase B-RAF. The present study thus explored whether B-RAF influences hERG channel expression and activity. To this end, hERG channels were expressed in Xenopus oocytes with or without wild-type B-RAF, hERG channel activity was determined utilizing dual-electrode voltage clamp and hERG protein abundance in the cell membrane was analyzed utilizing confocal microscopy as well as chemiluminescence. Moreover, in rhabdomyosarcoma RD cells the effect of B-RAF inhibitor PLX-4720 on hERG-mediated current was quantified by whole-cell patch clamp and hERG cell surface protein abundance by utilizing biotinylation of cell surface proteins as well as flow cytometry. As a result, co-expression of wild-type B-RAF in hERG-expressing Xenopus oocytes significantly increased hERG channel activity and hERG channel protein abundance in the cell membrane. Treatment for 24 hours of B-RAF and hERG-expressing Xenopus oocytes with B-RAF inhibitor PLX-4720 (10 µM) significantly decreased hERG-mediated current and hERG cell surface expression. Similarly, in rhabdomyosarcoma RD cells, treatment for 24 hours with B-RAF inhibitor PLX-4720 significantly decreased hERG cell membrane protein abundance and hERG-mediated current. In conclusion, B-RAF is a powerful regulator of hERG channel activity and cell surface hERG protein abundance.
Collapse
Affiliation(s)
| | | | - Ahmad Almilaji
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Aleksandra Lebedeva
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia
| | | | - Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
66
|
Liu J, Liu Y, Ren Y, Kang L, Zhang L. Transmembrane protein with unknown function 16A overexpression promotes glioma formation through the nuclear factor-κB signaling pathway. Mol Med Rep 2014; 9:1068-74. [PMID: 24401903 DOI: 10.3892/mmr.2014.1888] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/02/2014] [Indexed: 11/05/2022] Open
Abstract
Ion channels have been suggested to be important in the development and progression of tumors, however, chloride channels have rarely been analyzed in tumorigenesis. More recently, transmembrane protein with unknown function 16A (TMEM16A), hypothesized to be a candidate calcium‑activated Cl‑ channel, has been found to be overexpressed in a number of tumor types. Although several studies have implicated the overexpression of TMEM16A in certain tumor types, the exact role of TMEM16A in gliomas and the underlying mechanisms in tumorigenesis, remain poorly understood. In the present study, the role of TMEM16A in gliomas and the potential underlying mechanisms were analyzed. TMEM16A was highly abundant in various grades of gliomas and cultured glioma cells. Knockdown of TMEM16A suppressed cell proliferation, migration and invasion. Furthermore, nuclear factor‑κB (NF‑κB) was activated by overexpression of TMEM16A. In addition, TMEM16A regulated the expression of NF‑κB‑mediated genes, including cyclin D1, cyclin E and c‑myc, involved in cell proliferation, and matrix metalloproteinases (MMPs)‑2 and MMP‑9, which are associated with the migration and invasion of glioma cells. Collectively, results of the present study provide evidence for the involvement of TMEM16A in gliomas and the potential mechanism through which TMEM16A promotes glioma formation.
Collapse
Affiliation(s)
- Jun Liu
- Department of Geriatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yu Liu
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yingang Ren
- Department of Geriatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Li Kang
- Department of Geriatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Lihua Zhang
- Department of Geriatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
67
|
Abstract
Potassium channels are transmembrane proteins that selectively facilitate the flow of potassium ions down an electrochemical gradient. These molecules have been studied in great detail in the context of cell excitability, but their roles in less cell type-specific functions, such as cell proliferation, angiogenesis or cell migration, have only recently been assessed. Moreover, the importance of these channels for tumour biology has become evident. This, coupled with the fact that they are accessible proteins and that their pharmacology is well characterized, has increased the interest in investigating potassium channels as therapeutic targets in cancer patients.
Collapse
Affiliation(s)
- Luis A Pardo
- Oncophysiology Group, Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Walter Stühmer
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| |
Collapse
|
68
|
Crociani O, Zanieri F, Pillozzi S, Lastraioli E, Stefanini M, Fiore A, Fortunato A, D'Amico M, Masselli M, De Lorenzo E, Gasparoli L, Chiu M, Bussolati O, Becchetti A, Arcangeli A. hERG1 channels modulate integrin signaling to trigger angiogenesis and tumor progression in colorectal cancer. Sci Rep 2013; 3:3308. [PMID: 24270902 PMCID: PMC3839040 DOI: 10.1038/srep03308] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 11/05/2013] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a potential target for cancer therapy. We identified a novel signaling pathway that sustains angiogenesis and progression in colorectal cancer (CRC). This pathway is triggered by β1 integrin-mediated adhesion and leads to VEGF-A secretion. The effect is modulated by the human ether-à-go-go related gene 1 (hERG1) K(+) channel. hERG1 recruits and activates PI3K and Akt. This in turn increases the Hypoxia Inducible Factor (HIF)-dependent transcription of VEGF-A and other tumour progression genes. This signaling pathway has novel features in that the integrin- and hERG1-dependent activation of HIF (i) is triggered in normoxia, especially after CRC cells have experienced a hypoxic stage, (ii) involves NF-kB and (iii) is counteracted by an active p53. Blocking hERG1 switches this pathway off also in vivo, by inhibiting cell growth, angiogenesis and metastatic spread. This suggests that non-cardiotoxic anti-hERG1 drugs might be a fruitful therapeutic strategy to prevent the failure of anti-VEGF therapy.
Collapse
Affiliation(s)
- Olivia Crociani
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Francesca Zanieri
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Elena Lastraioli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Matteo Stefanini
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Antonella Fiore
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Angelo Fortunato
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Massimo D'Amico
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Marika Masselli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Emanuele De Lorenzo
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Luca Gasparoli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| | - Martina Chiu
- Unit of General Pathology Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT) University of Parma
| | - Ovidio Bussolati
- Unit of General Pathology Department of Biomedical, Biotechnological and Translational Sciences (SBiBiT) University of Parma
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano Bicocca, Milano, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence and Istituto Toscano Tumori (ITT), Florence, Italy
| |
Collapse
|
69
|
Filbin MG, Dabral SK, Pazyra-Murphy MF, Ramkissoon S, Kung AL, Pak E, Chung J, Theisen MA, Sun Y, Franchetti Y, Sun Y, Shulman DS, Redjal N, Tabak B, Beroukhim R, Wang Q, Zhao J, Dorsch M, Buonamici S, Ligon KL, Kelleher JF, Segal RA. Coordinate activation of Shh and PI3K signaling in PTEN-deficient glioblastoma: new therapeutic opportunities. Nat Med 2013; 19:1518-23. [PMID: 24076665 DOI: 10.1038/nm.3328] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 08/06/2013] [Indexed: 01/17/2023]
Abstract
In glioblastoma, phosphatidylinositol 3-kinase (PI3K) signaling is frequently activated by loss of the tumor suppressor phosphatase and tensin homolog (PTEN). However, it is not known whether inhibiting PI3K represents a selective and effective approach for treatment. We interrogated large databases and found that sonic hedgehog (SHH) signaling is activated in PTEN-deficient glioblastoma. We demonstrate that the SHH and PI3K pathways synergize to promote tumor growth and viability in human PTEN-deficient glioblastomas. A combination of PI3K and SHH signaling inhibitors not only suppressed the activation of both pathways but also abrogated S6 kinase (S6K) signaling. Accordingly, targeting both pathways simultaneously resulted in mitotic catastrophe and tumor apoptosis and markedly reduced the growth of PTEN-deficient glioblastomas in vitro and in vivo. The drugs tested here appear to be safe in humans; therefore, this combination may provide a new targeted treatment for glioblastoma.
Collapse
Affiliation(s)
- Mariella Gruber Filbin
- 1] Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA. [2] Department of Pediatric Oncology, Dana-Farber Cancer Institute and Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, USA. [3] Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA. [4] Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Fiore A, Carraresi L, Morabito A, Polvani S, Fortunato A, Lastraioli E, Femia AP, De Lorenzo E, Caderni G, Arcangeli A. Characterization of hERG1 channel role in mouse colorectal carcinogenesis. Cancer Med 2013; 2:583-94. [PMID: 24403225 PMCID: PMC3892791 DOI: 10.1002/cam4.72] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/29/2013] [Accepted: 02/14/2013] [Indexed: 12/13/2022] Open
Abstract
The human ether-à-go-go-related gene (hERG)1 K+ channel is upregulated in human colorectal cancer cells and primary samples. In this study, we examined the role of hERG1 in colorectal carcinogenesis using two mouse models: adenomatous polyposis coli (Apcmin/+) and azoxymethane (AOM)-treated mice. Colonic polyps of Apcmin/+ mice overexpressed mERG1 and their formation was reverted by the hERG1 blocker E4031. AOM was applied to either hERG1-transgenic (TG) mice, which overexpress hERG1 in the mucosa of the large intestine, or wild-type mice. A significant increase of both mucin-depleted foci and polyps in the colon of hERG1-TG mice was detected. Both the intestine of TG mice and colonic polyps of Apcmin/+ showed an upregulation of phospho-Protein Kinase B (pAkt)/vascular endothelial growth factor (VEGF-A) and an increased angiogenesis, which were reverted by treatment with E4031. On the whole, this article assigns a relevant role to hERG1 in the process of in vivo colorectal carcinogenesis.
Collapse
Affiliation(s)
- Antonella Fiore
- Department of Experimental and Clinical Medicine, Section of Internal Medicine and Oncology, University of Florence, Florence, Italy; Istituto Toscano Tumori, Florence, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Sphingosine 1-phosphate and cancer: lessons from thyroid cancer cells. Biomolecules 2013; 3:303-15. [PMID: 24970169 PMCID: PMC4030848 DOI: 10.3390/biom3020303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 02/03/2023] Open
Abstract
Sphingomyelin is found in the cell membrane of all eukaryotic cells, and was for a long time considered merely as a structural component. However, during the last two decades, metabolites of sphingomyelin, especially sphingosine 1-phosphate (S1P), have proven to be physiologically significant regulators of cell function. Through its five different G protein-coupled receptors, S1P regulates a wide array of cellular processes, ranging from stimulating cellular proliferation and migration, to the inhibition of apoptosis and induction of angiogenesis and modulation of cellular calcium homeostasis. Many of the processes regulated by S1P are important for normal cell physiology, but may also induce severe pathological conditions, especially in malignancies like cancer. Thus, understanding S1P signaling mechanisms has been the aim of a multitude of investigations. Great interest has also been shown in understanding the action of sphingosine kinase (SphK), i.e., the kinase phosphorylating sphingosine to S1P, and the interactions between S1P and growth factor signaling. In the present review, we will discuss recent findings regarding the possible importance of S1P and SphK in the etiology of thyroid cancer. Although clinical data is still scarce, our in vitro findings suggest that S1P may function as a “double-edged sword”, as the receptor profile of thyroid cancer cells largely determines whether S1P stimulates or blocks cellular migration. We will also discuss the interactions between S1P- and VEGF-evoked signaling, and the importance of a S1P1-VEGF receptor 2 complex in thyroid cancer cells.
Collapse
|
72
|
|
73
|
Li G, Hu Z, Yin H, Zhang Y, Huang X, Wang S, Li W. A novel dendritic nanocarrier of polyamidoamine-polyethylene glycol-cyclic RGD for "smart" small interfering RNA delivery and in vitro antitumor effects by human ether-à-go-go-related gene silencing in anaplastic thyroid carcinoma cells. Int J Nanomedicine 2013; 8:1293-306. [PMID: 23569377 PMCID: PMC3615931 DOI: 10.2147/ijn.s41555] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The application of RNA interference techniques is promising in gene therapeutic approaches, especially for cancers. To improve safety and efficiency of small interfering RNA (siRNA) delivery, a triblock dendritic nanocarrier, polyamidoamine-polyethylene glycol-cyclic RGD (PAMAM-PEG-cRGD), was developed and studied as an siRNA vector targeting the human ether-à-go-go-related gene (hERG) in human anaplastic thyroid carcinoma cells. Structure characterization, particle size, zeta potential, and gel retardation assay confirmed that complete triblock components were successfully synthesized with effective binding capacity of siRNA in this triblock nanocarrier. Cytotoxicity data indicated that conjugation of PEG significantly alleviated cytotoxicity when compared with unmodified PAMAM. PAMAM-PEG-cRGD exerted potent siRNA cellular internalization in which transfection efficiency measured by flow cytometry was up to 68% when the charge ratio (N/P ratio) was 3.5. Ligand-receptor affinity together with electrostatic interaction should be involved in the nano-siRNA endocytosis mechanism and we then proved that attachment of cRGD enhanced cellular uptake via RGD-integrin recognition. Gene silencing was evaluated by reverse transcription polymerase chain reaction and PAMAM-PEG-cRGD-siRNA complex downregulated the expression of hERG to 26.3% of the control value. Furthermore, gene knockdown of hERG elicited growth suppression as well as activated apoptosis by means of abolishing vascular endothelial growth factor secretion and triggering caspase-3 cascade in anaplastic thyroid carcinoma cells. Our study demonstrates that this novel triblock polymer, PAMAM-PEG-cRGD, exhibits negligible cytotoxicity, effective transfection, “smart” cancer targeting, and therefore is a promising siRNA nanocarrier.
Collapse
Affiliation(s)
- Guanhua Li
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
74
|
Role of HERG1 potassium channel in both malignant transformation and disease progression in head and neck carcinomas. Mod Pathol 2012; 25:1069-78. [PMID: 22460808 DOI: 10.1038/modpathol.2012.63] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Evidence indicates that human ether à-go-go-related gene 1 (HERG1) voltage-gated potassium channels could represent new valuable membrane therapeutic targets and diagnostic/prognostic biomarkers in various cancers. This study is the first to investigate the expression pattern of HERG1 potassium channel subunit in both primary tumors and precancerous lesions to establish its clinical and biological role during the development and progression of head and neck squamous cell carcinomas. HERG1 protein expression was evaluated by immunohistochemistry in paraffin-embedded tissue specimens from 133 patients with laryngeal/hypopharyngeal squamous cell carcinomas and 75 patients with laryngeal dysplasia, and correlated with clinical data. Our findings demonstrate that HERG1 is frequently aberrantly expressed in a high percentage of primary tumors (87%), whereas expression was negligible in both stromal cells and normal-adjacent epithelia. HERG1 expression increased during head and neck squamous cell carcinoma progression and was significantly associated with lymph node metastasis (P=0.04), advanced disease stages (P<0.001), regional tumor recurrence (P=0.004), distant metastasis (P=0.03) and reduced disease-specific survival (P=0.012, log-rank test). HERG1-positive expression was also detected in 31 (41%) of 75 laryngeal dysplasias. Interestingly, HERG1 expression increased with the grade of dysplasia; however, HERG1 expression but not histology correlated significantly with increased laryngeal cancer risk (P=0.007). In addition, functional studies in head and neck squamous cell carcinoma-derived cell lines further revealed that HERG1 expression promotes anchorage-dependent and -independent cell growth and invasive capability, although independently of its ion-conducting function. Our data demonstrate that HERG1 expression is a biologically and clinically relevant feature in head and neck squamous cell carcinoma progression and also during malignant transformation, and a promising candidate as cancer risk marker and therapeutic target for head and neck squamous cell carcinoma prevention and treatment.
Collapse
|
75
|
Njie-Mbye YF, Opere CA, Chitnis M, Ohia SE. Hydrogen sulfide: role in ion channel and transporter modulation in the eye. Front Physiol 2012; 3:295. [PMID: 22934046 PMCID: PMC3429066 DOI: 10.3389/fphys.2012.00295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/08/2012] [Indexed: 01/28/2023] Open
Abstract
Hydrogen sulfide (H2S), a colorless gas with a characteristic smell of rotten eggs, has been portrayed for decades as a toxic environmental pollutant. Since evidence of its basal production in mammalian tissues a decade ago, H2S has attracted substantial interest as a potential inorganic gaseous mediator with biological importance in cellular functions. Current research suggests that, next to its counterparts nitric oxide and carbon monoxide, H2S is an important multifunctional signaling molecule with pivotal regulatory roles in various physiological and pathophysiological processes as diverse as learning and memory, modulation of synaptic activities, cell survival, inflammation, and maintenance of vascular tone in the central nervous and cardiovascular systems. In contrast, there are few reports of a regulatory role of H2S in the eye. Accumulating reports on the pharmacological role of H2S in ocular tissues indicate the existence of a functional trans-sulfuration pathway and a potential physiological role for H2S as a gaseous neuromodulator in the eye. Thus, understanding the role of H2S in vision-related processes is imperative to our expanding knowledge of this molecule as a gaseous mediator in ocular tissues. This review aims to provide a comprehensive and current understanding of the potential role of H2S as a signaling molecule in the eye. This objective is achieved by discussing the involvement of H2S in the regulation of (1) ion channels such as calcium (L-type, T-type, and intracellular stores), potassium (KATP and small conductance channels) and chloride channels, (2) glutamate transporters such as EAAT1/GLAST and the L-cystine/glutamate antiporter. The role of H2S as an important mediator in cellular functions and physiological processes that are triggered by its interaction with ion channels/transporters in the eye will also be discussed.
Collapse
Affiliation(s)
- Ya F Njie-Mbye
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University Houston, TX, USA
| | | | | | | |
Collapse
|
76
|
Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG K+ Channels: Structure, Function, and Clinical Significance. Physiol Rev 2012; 92:1393-478. [DOI: 10.1152/physrev.00036.2011] [Citation(s) in RCA: 526] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The human ether-a-go-go related gene (hERG) encodes the pore-forming subunit of the rapid component of the delayed rectifier K+ channel, Kv11.1, which are expressed in the heart, various brain regions, smooth muscle cells, endocrine cells, and a wide range of tumor cell lines. However, it is the role that Kv11.1 channels play in the heart that has been best characterized, for two main reasons. First, it is the gene product involved in chromosome 7-associated long QT syndrome (LQTS), an inherited disorder associated with a markedly increased risk of ventricular arrhythmias and sudden cardiac death. Second, blockade of Kv11.1, by a wide range of prescription medications, causes drug-induced QT prolongation with an increase in risk of sudden cardiac arrest. In the first part of this review, the properties of Kv11.1 channels, including biogenesis, trafficking, gating, and pharmacology are discussed, while the second part focuses on the pathophysiology of Kv11.1 channels.
Collapse
Affiliation(s)
- Jamie I. Vandenberg
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Matthew D. Perry
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Mark J. Perrin
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Stefan A. Mann
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Ying Ke
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Adam P. Hill
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
77
|
Immunohistochemical biomarkers in gastric cancer research and management. Int J Surg Oncol 2012; 2012:868645. [PMID: 22778942 PMCID: PMC3388584 DOI: 10.1155/2012/868645] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/22/2012] [Accepted: 04/25/2012] [Indexed: 12/25/2022] Open
Abstract
Gastric cancer still represents a major health problem, despite a decrease in its incidence in the last years. Due to the social impact of gastric cancer (GC), there is a need for novel biomarkers in order to stratify patients into appropriate screening, surveillance, or treatment programs. Although histopathology remains the most reliable and less expensive method, numerous efforts have been made searching for novel biomarkers. In recent years, several molecules have been identified and tested for their clinical relevance in GC management. In this paper, we will focus on a well-known GC marker, whose determination is mandatory in GC, HER2, a marker whose correlation with prognosis is still controversial (VEGF-A) and a quite novel, unconventional marker, the ether-à-go-go-related gene 1 (hERG1). All these proteins can be easily detected with immunohistochemistry, a technique widely used both in diagnostic and research laboratories that represents a link between surgical and molecular pathology, basic science, and clinical medicine.
Collapse
|
78
|
Masselli M, Laise P, Tonini G, Fanelli D, Pillozzi S, Cetica V, Da Ros M, Sardi I, Buccoliero AM, Aricò M, Genitori L, Becchetti A, Arcangeli A. Deregulation of ion channel and transporter encoding genes in pediatric gliomas. Front Oncol 2012; 2:53. [PMID: 22662319 PMCID: PMC3362739 DOI: 10.3389/fonc.2012.00053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 05/08/2012] [Indexed: 02/05/2023] Open
Abstract
Brain tumors, including the majority gliomas, are the leading cause of cancer-related death in children. World Health Organization has divided pediatric brain tumors into different grades and, based upon cDNA microarray data identifying gene expression profiles (GEPs), it has become evident in the last decade that the various grades involve different types of genetic alterations. However, it is not known whether ion channel and transporter genes, intimately involved in brain functioning, are associated with such GEPs. We determined the GEPs in an available cohort of 10 pediatric brain tumors initially by comparing the data obtained from four primary tumor samples and corresponding short-term cultures. The correspondence between the two types of samples was statistically significant. We then performed bioinformatic analyses on those samples (a total of nine) which corresponded to tumors of glial origin, either tissues or cell cultures, depending on the best "RNA integrity number." We used R software to evaluate the genes which were differentially expressed (DE) in gliomas compared with normal brain. Applying a p-value below 0.01 and fold change ≥4, led to identification of 2284 DE genes. Through a Functional Annotation Analysis (FAA) using the NIH-DAVID software, the DE genes turned out to be associated mainly with: immune/inflammatory response, cell proliferation and survival, cell adhesion and motility, neuronal phenotype, and ion transport. We have shown that GEPs of pediatric brain tumors can be studied using either primary tumor samples or short-term cultures with similar results. From FAA, we concluded that, among DE genes, pediatric gliomas show a strong deregulation of genes related to ion channels and transporters.
Collapse
Affiliation(s)
- Marika Masselli
- Department of Experimental Pathology and Oncology, University of Firenze Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Lastraioli E, Bencini L, Bianchini E, Romoli MR, Crociani O, Giommoni E, Messerini L, Gasperoni S, Moretti R, Di Costanzo F, Boni L, Arcangeli A. hERG1 Channels and Glut-1 as Independent Prognostic Indicators of Worse Outcome in Stage I and II Colorectal Cancer: A Pilot Study. Transl Oncol 2012; 5:105-112. [PMID: 22496927 PMCID: PMC3323932 DOI: 10.1593/tlo.11250] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 12/06/2011] [Accepted: 12/07/2011] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND There is a need to identify new markers to assess recurrence risk in early-stage colorectal cancer (CRC) patients. We explored the prognostic impact of ether-a-gò-gò-related gene 1 channels and some hypoxia markers, in patients with nonmetastatic (stage I, II, and III) CRC. METHODS The expression of hERG1, vascular endothelial growth factor A (VEGF-A), glucose transporter 1, carbonic anhydrase IX (CA-IX), epidermal growth factor receptor (EGF-R), and p53 was tested by immunohistochemistry in 135 patients. The median follow-up was 35 months. Clinicopathologic parameters and overall survival were evaluated. RESULTS hERG1 displayed a statistically significant association with Glut-1, VEGF-A, CA-IX, and EGF-R; p53 with VEGF-A and CA-IX; Glut-1 with the age of the patients; and EGF-R with TNM and mucin content. TNM and CA-IX were prognostic factors at the univariate analysis; TNM, hERG1, and Glut-1, at the multivariate analysis. Risk scores calculated from the final multivariate model allowed to stratify patients into four different risk groups: A) stage I-II, Glut-1 positivity, any hERG1; B) stage I-II, Glut-1 and hERG1 negativity; C) stage I-II, Glut-1 negativity, hERG1 positivity; D) stage III, any Glut-1 and any hERG1. CONCLUSIONS hERG1 positivity with Glut-1 negativity identifies a patient group with poor prognosis within stage I-II CRC. The possibility that these patients might benefit from adjuvant therapy, independently from the TNM stage, is discussed. IMPACT More robust prognostic and predictive markers, supplementing standard clinical and pathologic staging, are needed for node-negative patients.
Collapse
Affiliation(s)
- Elena Lastraioli
- Department of Experimental Pathology and Oncology, University of Florence, Istituto Toscano Tumori, Florence, Italy
| | - Lapo Bencini
- General Surgery and Surgical Oncology, Azienda Ospedaliero-Universitaria, Careggi, Florence, Italy
| | - Elisa Bianchini
- Clinical Trials Coordinating Center, Azienda Ospedaliero-Universitaria, Careggi, Florence, Italy
| | - Maria Raffaella Romoli
- Department of Experimental Pathology and Oncology, University of Florence, Istituto Toscano Tumori, Florence, Italy
| | - Olivia Crociani
- Department of Experimental Pathology and Oncology, University of Florence, Istituto Toscano Tumori, Florence, Italy
| | - Elisa Giommoni
- Medical Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Luca Messerini
- Department of Human Pathology and Oncology, University of Florence, Florence, Italy
| | - Silvia Gasperoni
- Medical Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Renato Moretti
- General Surgery and Surgical Oncology, Azienda Ospedaliero-Universitaria, Careggi, Florence, Italy
| | | | - Luca Boni
- Clinical Trials Coordinating Center, Azienda Ospedaliero-Universitaria, Careggi, Florence, Italy
| | - Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Florence, Istituto Toscano Tumori, Florence, Italy
| |
Collapse
|
80
|
Yarotskyy V, Elmslie KS. Roscovitine inhibits CaV3.1 (T-type) channels by preferentially affecting closed-state inactivation. J Pharmacol Exp Ther 2012; 340:463-72. [PMID: 22088954 PMCID: PMC3263959 DOI: 10.1124/jpet.111.187104] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/15/2011] [Indexed: 12/18/2022] Open
Abstract
T-type calcium channels (Ca(V)3) play an important role in many physiological and pathological processes, including cancerogenesis. Ca(V)3 channel blockers have been proposed as potential cancer treatments. Roscovitine, a trisubstituted purine, is a cyclin-dependent kinase (CDK) inhibitor that is currently undergoing phase II clinical trials as an anticancer drug and has been shown to affect calcium and potassium channel activity. Here, we investigate the effect of roscovitine on Ca(V)3.1 channels. Ca(V)3.1 channels were transiently expressed in human embryonic kidney 293 cells, and currents were recorded by using the whole-cell patch-clamp technique. Roscovitine blocks Ca(V)3.1 channels with higher affinity for depolarized cells (EC₅₀ of 10 μM), which is associated with a negative shift in the voltage dependence of closed-state inactivation. Enhanced inactivation is mediated by roscovitine-induced acceleration of closed-state inactivation and slowed recovery from inactivation. Small effects of roscovitine were also observed on T-channel deactivation and open-state inactivation, but neither could explain the inhibitory effect. Roscovitine inhibits Ca(V)3.1 channels within the therapeutic range (10-50 μM) in part by stabilizing the closed-inactivated state. The ability of roscovitine to block multiple mediators of proliferation, including CDKs and Ca(V)3.1 channels, may facilitate its anticancer properties.
Collapse
Affiliation(s)
- Viktor Yarotskyy
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | |
Collapse
|
81
|
Zheng F, Li H, Liang K, Du Y, Guo D, Huang S. Imatinib has the potential to exert its antileukemia effects by down-regulating hERG1 K+ channels in chronic myelogenous leukemia. Med Oncol 2011; 29:2127-35. [PMID: 22161019 DOI: 10.1007/s12032-011-0102-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 10/27/2011] [Indexed: 11/25/2022]
Abstract
Imatinib is a powerful protein tyrosine kinase (PTK) inhibitor that specifically targets BCR-ABL, KIT, and PDGFR kinases, has become the current first-line therapy for all newly diagnosed chronic myeloid leukemia (CML). Beside PTKs, PTK inhibitors alter the activity of a large number of voltage-dependent ion channels. hERG1 K(+) channels are highly expressed in leukemia cells and appear of exceptional importance in favoring leukemogenesis. The present study explored a possible regulatory effect of imatinib upon hERG1 K(+) channels as a means to uncover new molecular events involved in the antileukemic activity of this PTK inhibitor in CML. The results demonstrated that hERG1 was highly detected in K562 cells and primary CML cells, and down-regulated by imatinib at mRNA and protein levels. Furthermore, imatinib markedly reduced hERG currents in HEK293T-hERG cells, this effect was accompanied by inhibition of CML cell proliferation and apoptosis, as well as suppression of vascular endothelial growth factor (VEGF) secretion. Moreover, these antileukemia effects of imatinib were potentiated by E-4031, a specific hERG1 inhibitor. Together, these results provide evidence of a novel potential molecular mechanism of antileukemic activities by imatinib which, independent of targeting tyrosine kinase, highlight hERG1 K(+) channels as a therapeutic target for CML treatment.
Collapse
Affiliation(s)
- Fang Zheng
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1277, Wuhan 430022, People's Republic of China
| | | | | | | | | | | |
Collapse
|
82
|
Molenaar RJ. Ion channels in glioblastoma. ISRN NEUROLOGY 2011; 2011:590249. [PMID: 22389824 PMCID: PMC3263536 DOI: 10.5402/2011/590249] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 09/19/2011] [Indexed: 11/23/2022]
Abstract
Glioblastoma is the most common primary brain tumor with the most dismal prognosis. It is characterized by extensive invasion, migration, and angiogenesis. Median survival is only 15 months due to this behavior, rendering focal surgical resection ineffective and adequate radiotherapy impossible. At this moment, several ion channels have been implicated in glioblastoma proliferation, migration, and invasion. This paper summarizes studies on potassium, sodium, chloride, and calcium channels of glioblastoma. It provides an up-to-date overview of the literature that could ultimately lead to new therapeutic targets.
Collapse
Affiliation(s)
- Remco J Molenaar
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
83
|
Glassmeier G, Hempel K, Wulfsen I, Bauer CK, Schumacher U, Schwarz JR. Inhibition of HERG1 K+ channel protein expression decreases cell proliferation of human small cell lung cancer cells. Pflugers Arch 2011; 463:365-76. [PMID: 22075718 PMCID: PMC3261411 DOI: 10.1007/s00424-011-1045-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/12/2011] [Accepted: 10/14/2011] [Indexed: 01/30/2023]
Abstract
HERG (human ether-à-go-go-related gene) K+ currents fulfill important ionic functions in cardiac and other excitable cells. In addition, HERG channels influence cell growth and migration in various types of tumor cells. The mechanisms underlying these functions are still not resolved. Here, we investigated the role of HERG channels for cell growth in a cell line (SW2) derived from small cell lung cancer (SCLC), a malignant variant of lung cancer. The two HERG1 isoforms (HERG1a, HERG1b) as well as HERG2 and HERG3 are expressed in SW2 cells. Inhibition of HERG currents by acute or sustained application of E-4031, a specific ERG channel blocker, depolarized SW2 cells by 10–15 mV. This result indicated that HERG K+ conductance contributes considerably to the maintenance of the resting potential of about −45 mV. Blockage of HERG channels by E-4031 for up to 72 h did not affect cell proliferation. In contrast, siRNA-induced inhibition of HERG1 protein expression decreased cell proliferation by about 50%. Reduction of HERG1 protein expression was confirmed by Western blots. HERG current was almost absent in SW2 cells transfected with siRNA against HERG1. Qualitatively similar results were obtained in three other SCLC cell lines (OH1, OH3, H82), suggesting that the HERG1 channel protein is involved in SCLC cell growth, whereas the ion-conducting function of HERG1 seems not to be important for cell growth.
Collapse
Affiliation(s)
- Günter Glassmeier
- Institut für Zelluläre und Integrative Physiologie, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Martinistr. 52, D-20246, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
84
|
Arcangeli A. Ion channels and transporters in cancer. 3. Ion channels in the tumor cell-microenvironment cross talk. Am J Physiol Cell Physiol 2011; 301:C762-71. [DOI: 10.1152/ajpcell.00113.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The traditional view of cancer as a collection of proliferating cells must be reconsidered, and cancer must be viewed as a “tissue” constituted by both transformed cells and a heterogeneous microenvironment, that tumor cells construct and remodel during multistep tumorigenesis. The “tumor microenvironment” (TM) is formed by mesenchymal, endothelial, and immune cells immersed in a network of extracellular matrix (ECM) proteins and soluble factors. The TM strongly contributes to tumor progression, through long distance, cell-to-cell or cell-to-matrix signals, which influence different aspects of tumor cell behavior. Understanding the relationships among the different components of the cancer tissue is crucial to design and develop new therapeutic strategies. Ion channels are emerging as relevant players in the cross talk between tumor cells and their TM. Ion channels are expressed on tumor cells, as well as in the different cellular components of the TM. In all these cells, ion channels are in a strategic position to sense and transmit extracellular signals into the intracellular machinery. Often, this transmission is mediated by integrin adhesion receptors, which can be functional partners of ion channels since they form molecular complexes with the channel protein in the context of the plasma membrane. The same relevant role is exerted by ion transporters, which also contribute to determine two facets of the cancer tissue: hypoxia and the acidic extracellular pH. On the whole, it is conceivable to prospect the targeting of ion channels for new therapeutic strategies aimed at better controlling the malignant progression of the cancer tissue.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Firenze, and Istituto Toscano Tumori, Firenze, Italy
| |
Collapse
|
85
|
Abstract
The human ether-a-go-go-related gene potassium channel (hERG, Kv11.1, KCNH2) has an essential role in cardiac action potential repolarization. Electrical dysfunction of the voltage-sensitive ion channel is associated with potentially lethal ventricular arrhythmias in humans. hERG K+ channels are also expressed in a variety of cancer cells where they control cell proliferation and apoptosis. In this review, we discuss molecular mechanisms of hERG-associated cell cycle regulation and cell death. In addition, the significance of hERG K+ channels as future drug target in anticancer therapy is highlighted.
Collapse
|
86
|
Becchetti A. Ion channels and transporters in cancer. 1. Ion channels and cell proliferation in cancer. Am J Physiol Cell Physiol 2011; 301:C255-65. [DOI: 10.1152/ajpcell.00047.2011] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Progress through the cell mitotic cycle requires precise timing of the intrinsic molecular steps and tight coordination with the environmental signals that maintain a cell into the proper physiological context. Because of their great functional flexibility, ion channels coordinate the upstream and downstream signals that converge on the cell cycle machinery. Both voltage- and ligand-gated channels have been implicated in the control of different cell cycle checkpoints in normal as well as neoplastic cells. Ion channels mediate the calcium signals that punctuate the mitotic process, the cell volume oscillations typical of cycling cells, and the exocytosis of autocrine or angiogenetic factors. Other functions of ion channels in proliferation are still matter of debate. These may or may not depend on ion transport, as the channel proteins can form macromolecular complexes with growth factor and cell adhesion receptors. Direct conformational coupling with the cytoplasmic regulatory proteins is also possible. Derangement or relaxed control of the above processes can promote neoplasia. Specific types of ion channels have turned out to participate in the different stages of the tumor progression, in which cell heterogeneity is increased by the selection of malignant cell clones expressing the ion channel types that better support unrestrained growth. However, a comprehensive mechanistic picture of the functional relations between ion channels and cell proliferation is yet not available, partly because of the considerable experimental challenges offered by studying these processes in living mammalian cells. No doubt, such studies will constitute one of the most fruitful research fields for the next generation of cell physiologists.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
87
|
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
|
88
|
Wang Y, Zhang Y, Yang L, Cai B, Li J, Zhou Y, Yin L, Yang L, Yang BF, Lu YJ. Arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells through activation of caspase-3 and inhibition of HERG channels. Exp Ther Med 2011; 2:481-486. [PMID: 22977528 DOI: 10.3892/etm.2011.224] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 02/28/2011] [Indexed: 01/27/2023] Open
Abstract
Arsenic trioxide (As(2)O(3)) has been widely used to treat patients with acute promyelocytic leukemia and has also been shown to exhibit therapeutic effects on various types of solid tumors, including gastric cancer and lung carcinoma. Breast cancer is a type of solid tumor whose incidence has been increasing for many years. The present study was designed to investigate the effects of As(2)O(3) on the human breast cancer cell line MCF-7, and to explore its potential mechanisms. The MTT assay demonstrated that As(2)O(3) decreased the cellular viability of MCF-7 cells in a concentration-dependent manner. Morphological observation, the TUNEL assay and flow cytometric analysis revealed that apoptosis was involved in the process. An assay for caspase-3 activity suggested that the apoptosis was mediated through caspase-3 activation. Further investigation indicated that protein levels of the human ether-a-go-go-related gene (HERG) were markedly downregulated by As(2)O(3). Taken together, the results indicate that arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells at least in part through the activation of caspase-3 and the decrease in HERG expression.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine and Pharmaceutics, and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Pillozzi S, Masselli M, De Lorenzo E, Accordi B, Cilia E, Crociani O, Amedei A, Veltroni M, D'Amico M, Basso G, Becchetti A, Campana D, Arcangeli A. Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers. Blood 2011; 117:902-914. [PMID: 21048156 DOI: 10.1182/blood-2010-01-262691] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Bone marrow mesenchymal cells (MSCs) can protect leukemic cells from chemotherapy, thus increasing their survival rate. We studied the potential molecular mechanisms underlying this effect in acute lymphoblastic leukemia (ALL) cells. Coculture of ALL cells with MSCs induced on the lymphoblast plasma membrane the expression of a signaling complex formed by hERG1 (human ether-à-go-go-related gene 1) channels, the β(1)-integrin subunit, and the chemokine receptor CXC chemokine receptor-4. The assembly of such a protein complex activated both the extracellular signal-related kinase 1/2 (ERK1/2) and the phosphoinositide 3-kinase (PI3K)/Akt prosurvival signaling pathways. At the same time, ALL cells became markedly resistant to chemotherapy-induced apoptosis. hERG1 channel function appeared to be important for both the initiation of prosurvival signals and the development of drug resistance, because specific channel blockers decreased the protective effect of MSCs. NOD/SCID mice engrafted with ALL cells and treated with channel blockers showed reduced leukemic infiltration and had higher survival rates. Moreover, hERG1 blockade enhanced the therapeutic effect produced by corticosteroids. Our findings provide a rationale for clinical testing of hERG1 blockers in the context of antileukemic therapy for patients with ALL.
Collapse
Affiliation(s)
- Serena Pillozzi
- Department of Experimental Pathology and Oncology, University of Firenze, Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Jie S, Li H, Tian Y, Guo D, Zhu J, Gao S, Jiang L. Berberine inhibits angiogenic potential of Hep G2 cell line through VEGF down-regulation in vitro. J Gastroenterol Hepatol 2011; 26:179-85. [PMID: 21175812 DOI: 10.1111/j.1440-1746.2010.06389.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Berberine, an herbal alkaloid, has been reported to have promotion potential of apoptosis and anticancer effect on a variety of human tumor cells. To obtain more specific understanding of those consequences of berberine on hepatocellular carcinoma (HCC) and the tumor microenvironment, we conducted in vitro experiments to investigate the inhibitory effect of berberine on tumor-induced angiogenesis using HCC cells and human umbilical vein endothelial cells (HUVECs). METHODS Human umbilical vein endothelial cell growth was quantified with the CCK-8 cell proliferation assay; cell migration was observed with a Boyden chamber (Transwell, Corning, Lowell, MA, USA), and angiogenesis was assessed by endothelial tube formation in Matrigel in vitro. In addition, VEGF level was determined by ELISA and VEGF mRNA expression by RT-PCR. RESULTS Berberine inhibited the capacity of HCC to stimulate HUVEC's proliferation, migration and endothelial tube formation, suggesting that berberine could influence the cross-talk between the HCC cell and vascular endothelial cells. These results demonstrate berberine's antiangiogenesis property and its clinical potential as an inhibitor of tumor angiogenesis. Subsequently analyses reveal that berberine prevents secretion of VEGF from HCC and down-regulates VEGF mRNA expression. CONCLUSION These findings strongly suggest that berberine is a potential antiangiogenic agent and a promising antitumor drug for HCC.
Collapse
Affiliation(s)
- Shenghua Jie
- Department of Infectious Diseases, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
91
|
Asher V, Sowter H, Shaw R, Bali A, Khan R. Eag and HERG potassium channels as novel therapeutic targets in cancer. World J Surg Oncol 2010; 8:113. [PMID: 21190577 PMCID: PMC3022597 DOI: 10.1186/1477-7819-8-113] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/29/2010] [Indexed: 12/03/2022] Open
Abstract
Voltage gated potassium channels have been extensively studied in relation to cancer. In this review, we will focus on the role of two potassium channels, Ether à-go-go (Eag), Human ether à-go-go related gene (HERG), in cancer and their potential therapeutic utility in the treatment of cancer. Eag and HERG are expressed in cancers of various organs and have been implicated in cell cycle progression and proliferation of cancer cells. Inhibition of these channels has been shown to reduce proliferation both in vitro and vivo studies identifying potassium channel modulators as putative inhibitors of tumour progression. Eag channels in view of their restricted expression in normal tissue may emerge as novel tumour biomarkers.
Collapse
Affiliation(s)
- Viren Asher
- Department of Obstetrics and Gynaecology, School of Graduate Medicine and Health, Royal Derby Hospital, Uttoxeter road, Derby DE22 3DT, UK.
| | | | | | | | | |
Collapse
|
92
|
Integrins and ion channels in cell migration: implications for neuronal development, wound healing and metastatic spread. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 674:107-23. [PMID: 20549944 DOI: 10.1007/978-1-4419-6066-5_10] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cells migration is necessary for proper embryonic development and adult tissue remodeling. Its mechanisms determine the physiopathology of processes such as neuronal targeting, inflammation, wound healing and metastatic spread. Crawling of cells onto solid surfaces requires a controlled sequence of cell protrusions and retractions that mainly depends on sophisticated regulation of the actin cytoskeleton, although the contribution of microtubules should not be neglected. This process is triggered and modulated by a combination of diffusible and fixed environmental signals. External cues are sensed and integrated by membrane receptors, including integrins, which transduce these signals into cellular signaling pathways, often centered on the small GTPase proteins belonging to the Rho family. These pathways regulate the coordinated cytoskeletal rearrangements necessary for proper timing of adhesion, contraction and detachement at the front and rear side of cells finding their way through the extracellular spaces. The overall process involves continuous modulation of cell motility, shape and volume, in which ion channels play major roles. In particular, Ca2+ signals have both global and local regulatory effects on cell motility, because they target the contractile proteins as well as many regulatory proteins. After reviewing the fundamental mechanisms of eukaryotic cell migration onto solid substrates, we briefly describe how integrin receptors and ion channels are involved in cell movement. We next examine a few processes in which these mechanisms have been studied in depth. We thus illustrate how integrins and K+ channels control cell volume and migration, how intracellular Ca2+ homeostasis affects the motility of neuronal growth cones and what is known about the ion channel roles in epithelial cell migration. These mechanisms are implicated in a variety of pathological processes, such as the disruption of neural circuits and wound healing. Finally, we describe the interaction between neoplastic cells and their local environment and how derangement of adhesion can lead to metastatic spread. It is likely that the cellular mechanisms controlled by integrin receptors, ion channels or both participate in the entire metastatic process. Until now, however, evidence is limited to a few steps of the metastatic cascade, such as brain tumor invasiveness.
Collapse
|
93
|
Arcangeli A, Becchetti A. New Trends in Cancer Therapy: Targeting Ion Channels and Transporters. Pharmaceuticals (Basel) 2010; 3:1202-1224. [PMID: 27713296 PMCID: PMC4034029 DOI: 10.3390/ph3041202] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 03/25/2010] [Accepted: 03/29/2010] [Indexed: 02/07/2023] Open
Abstract
The expression and activity of different channel types mark and regulate specific stages of cancer establishment and progression. Blocking channel activity impairs the growth of some tumors, both in vitro and in vivo, which opens a new field for pharmaceutical research. However, ion channel blockers may produce serious side effects, such as cardiac arrhythmias. For instance, Kv11.1 (hERG1) channels are aberrantly expressed in several human cancers, in which they control different aspects of the neoplastic cell behaviour. hERG1 blockers tend to inhibit cancer growth. However they also retard the cardiac repolarization, thus lengthening the electrocardiographic QT interval, which can lead to life-threatening ventricular arrhythmias. Several possibilities exist to produce less harmful compounds, such as developing specific drugs that bind hERG1 channels in the open state or disassemble the ion channel/integrin complex which appears to be crucial in certain stages of neoplastic progression. The potential approaches to improve the efficacy and safety of ion channel targeting in oncology include: (1) targeting specific conformational channel states; (2) finding ever more specific inhibitors, including peptide toxins, for channel subtypes mainly expressed in well-identified tumors; (3) using specific ligands to convey traceable or cytotoxic compounds; (4) developing channel blocking antibodies; (5) designing new molecular tools to decrease channel expression in selected cancer types. Similar concepts apply to ion transporters such as the Na⁺/K⁺ pump and the Na⁺/H⁺ exchanger. Pharmacological targeting of these transporters is also currently being considered in anti-neoplastic therapy.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Florence, Italy.
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy.
| | - Andrea Becchetti
- Department of Experimental Pathology and Oncology, University of Florence, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| |
Collapse
|
94
|
Ganapathi SB, Fox TE, Kester M, Elmslie KS. Ceramide modulates HERG potassium channel gating by translocation into lipid rafts. Am J Physiol Cell Physiol 2010; 299:C74-86. [PMID: 20375276 DOI: 10.1152/ajpcell.00462.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Human ether-à-go-go-related gene (HERG) potassium channels play an important role in cardiac action potential repolarization, and HERG dysfunction can cause cardiac arrhythmias. However, recent evidence suggests a role for HERG in the proliferation and progression of multiple types of cancers, making it an attractive target for cancer therapy. Ceramide is an important second messenger of the sphingolipid family, which due to its proapoptotic properties has shown promising results in animal models as an anticancer agent. Yet the acute effects of ceramide on HERG potassium channels are not known. In the present study we examined the effects of cell-permeable C(6)-ceramide on HERG potassium channels stably expressed in HEK-293 cells. C(6)-ceramide (10 microM) reversibly inhibited HERG channel current (I(HERG)) by 36 +/- 5%. Kinetically, ceramide induced a significant hyperpolarizing shift in the current-voltage relationship (DeltaV(1/2) = -8 +/- 0.5 mV) and increased the deactivation rate (43 +/- 3% for tau(fast) and 51 +/- 3% for tau(slow)). Mechanistically, ceramide recruited HERG channels within caveolin-enriched lipid rafts. Cholesterol depletion and repletion experiments and mathematical modeling studies confirmed that inhibition and gating effects are mediated by separate mechanisms. The ceramide-induced hyperpolarizing gating shift (raft mediated) could offset the impact of inhibition (raft independent) during cardiac action potential repolarization, so together they may nullify any negative impact on cardiac rhythm. Our results provide new insights into the effects of C(6)-ceramide on HERG channels and suggest that C(6)-ceramide can be a promising therapeutic for cancers that overexpress HERG.
Collapse
Affiliation(s)
- Sindura B Ganapathi
- Department of Pharmacology, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
95
|
Ding XW, Yang WB, Gao S, Wang W, Li Z, Hu WM, Li JJ, Luo HS. Prognostic significance of hERG1 expression in gastric cancer. Dig Dis Sci 2010; 55:1004-10. [PMID: 19495974 DOI: 10.1007/s10620-009-0834-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 04/30/2009] [Indexed: 01/12/2023]
Abstract
Previous studies have demonstrated that human ether-à-go-go-related potassium channel (hERG1) is highly expressed in many tumor cell lines, as well as in primary human cancers, and, hence, have a critical role in cell cycle progress and proliferation. In this study, hERG1 expression was investigated in gastric cancer by immunohistochemistry and/or reverse transcription polymerase chain reaction (RT-PCR). It was discovered that hERG1, which was negatively expressed in surrounding non-tumor tissues, switched to aberrantly positive expression in gastric cancer. Statistically, there were significant differences in hERG1 protein expression according to factors such as serosal invasion, venous invasion, lymph node metastases, other organ metastases, and stage. The mean survival time for the hERG1-positive expression group was significantly shorter than the negative group, the survival rates for the positive group were significantly lower than the negative group, and hERG1 expression was found to be an independent prognostic factor. In summary, hERG1 channel was proved to be a potential biomarker for gastric cancer invasion and survival.
Collapse
Affiliation(s)
- Xiang-Wu Ding
- Department of Gastroenterology, Xiangfan Central Hospital, Xiangfan, 441021, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Physical and Functional Interaction between Integrins and hERG1 Channels in Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 674:55-67. [DOI: 10.1007/978-1-4419-6066-5_6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
97
|
Abstract
The human genome encodes 40 voltage-gated K(+) channels (K(V)), which are involved in diverse physiological processes ranging from repolarization of neuronal and cardiac action potentials, to regulating Ca(2+) signalling and cell volume, to driving cellular proliferation and migration. K(V) channels offer tremendous opportunities for the development of new drugs to treat cancer, autoimmune diseases and metabolic, neurological and cardiovascular disorders. This Review discusses pharmacological strategies for targeting K(V) channels with venom peptides, antibodies and small molecules, and highlights recent progress in the preclinical and clinical development of drugs targeting the K(V)1 subfamily, the K(V)7 subfamily (also known as KCNQ), K(V)10.1 (also known as EAG1 and KCNH1) and K(V)11.1 (also known as HERG and KCNH2) channels.
Collapse
|
98
|
Cui G, Shu W, Wu Q, Chen Y. Effect of Gambogic acid on the regulation of hERG channel in K562 cells in vitro. ACTA ACUST UNITED AC 2009; 29:540-5. [DOI: 10.1007/s11596-009-0503-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Indexed: 02/07/2023]
|
99
|
The role of hERG1 K+ channels and a functional link between hERG1 K+ channels and SDF-1 in acute leukemic cell migration. Exp Cell Res 2009; 315:2256-64. [DOI: 10.1016/j.yexcr.2009.04.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 04/13/2009] [Accepted: 04/21/2009] [Indexed: 11/22/2022]
|
100
|
Li H, Liu L, Guo L, Zhang J, Du W, Li X, Liu W, Chen X, Huang S. HERG K+ channel expression in CD34+/CD38-/CD123(high) cells and primary leukemia cells and analysis of its regulation in leukemia cells. Int J Hematol 2009; 87:387-392. [PMID: 18415658 DOI: 10.1007/s12185-008-0056-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 01/05/2008] [Accepted: 01/25/2008] [Indexed: 12/30/2022]
Abstract
The human ether-a-go-go-related (herg) gene encoding K+ channels (HERG) belongs to an evolutionarily conserved multigene family of voltage activated K+ channels. The functional properties of HERG K+ channels are complex and their contribution to the repolarization of the cardiac action potential are well understood. Recent studies revealed that HERG K+ channels are preferentially expressed in different histogenesis of tumor cells. Leukemia is a cancer that originates in the bone marrow hematopoietic stem cells (HSCs). Leukemia stem cells (LSCs) are critical in the perpetuation of the disease. A better understanding of LSCs and molecular biology will allow the design of more effective therapies. We report in this study that herg was expressed in CD34+/CD38-/ CD123(high) LSCs but not expressed in normal bone marrow CD34+/CD38- HSCs. In addtion, herg is also expressed in leukemia cell lines K562 and HL-60 and almost all the primary leukemia cells whereas not in the normal bone marrow cells. In addition, the expression of herg mRNA was not associated with the clinical and cytogenetic feature of leukemia. Moreover, HERG K+ channels can regulate leukemia cells proliferation and cell cycle. These data provide evidence for the oncogenic potential of HERG K+ channels and it may be a novel, potential pharmacological target for leukemia therapy in the future.
Collapse
Affiliation(s)
- Huiyu Li
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Liqiong Liu
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Linlin Guo
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Jiahua Zhang
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Wen Du
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Xiaoqing Li
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Wei Liu
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Xiangjun Chen
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China
| | - Shiang Huang
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiegang Avenue, 430022, Wuhan, China.
| |
Collapse
|