51
|
Martínez R, Stühmer W, Martin S, Schell J, Reichmann A, Rohde V, Pardo L. Analysis of the expression of Kv10.1 potassium channel in patients with brain metastases and glioblastoma multiforme: impact on survival. BMC Cancer 2015; 15:839. [PMID: 26530050 PMCID: PMC4632660 DOI: 10.1186/s12885-015-1848-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/26/2015] [Indexed: 01/01/2023] Open
Abstract
Background Kv10.1, a voltage-gated potassium channel only detected in the healthy brain, was found to be aberrantly expressed in extracerebral cancers. Investigations of Kv10.1 in brain metastasis and glioblastoma multiforme (GBM) are lacking. Methods We analyzed the expression of Kv10.1 by immunohistochemistry in these brain tumors (75 metastasis from different primary tumors, 71 GBM patients) and the influence of a therapy with tricyclic antidepressants (which are Kv10.1 blockers) on survival. We also investigated Kv10.1 expression in the corresponding primary carcinomas of metastases patients. Results We observed positive Kv10.1 expression in 85.3 % of the brain metastases and in 77.5 % of GBMs. Patients with brain metastases, showing low Kv10.1 expression, had a significantly longer overall survival compared to those patients with high Kv10.1 expression. Metastases patients displaying low Kv10.1 expression and also receiving tricyclic antidepressants showed a significantly longer median overall survival as compared to untreated patients. Conclusions Our data show that Kv10.1 is not only highly expressed in malignant tumors outside CNS, but also in the most frequent cerebral cancer entities, metastasis and GBM, which remain incurable in spite of aggressive multimodal therapies. Our results extend the correlation between dismal prognosis and Kv10.1 expression to patients with brain metastases or GBMs and, moreover, they strongly suggest a role of tricyclic antidepressants for personalized therapy of brain malignancies.
Collapse
Affiliation(s)
- Ramón Martínez
- Department of Neurosurgery, University of Goettingen, Robert-Koch-Str. 40, Goettingen, 37075, Germany.
| | - Walter Stühmer
- Department of Molecular Biology of Neuronal Signals, Max-Planck Institute for Experimental Medicine, Hermann-Rein-Str. 3, Goettingen, 37075, Germany.
| | - Sabine Martin
- Department of Molecular Biology of Neuronal Signals, Max-Planck Institute for Experimental Medicine, Hermann-Rein-Str. 3, Goettingen, 37075, Germany.
| | - Julian Schell
- Department of Neurosurgery, University of Goettingen, Robert-Koch-Str. 40, Goettingen, 37075, Germany.
| | - Andrea Reichmann
- Department of Neurosurgery, University of Goettingen, Robert-Koch-Str. 40, Goettingen, 37075, Germany.
| | - Veit Rohde
- Department of Neurosurgery, University of Goettingen, Robert-Koch-Str. 40, Goettingen, 37075, Germany.
| | - Luis Pardo
- Department of Molecular Biology of Neuronal Signals, Max-Planck Institute for Experimental Medicine, Hermann-Rein-Str. 3, Goettingen, 37075, Germany.
| |
Collapse
|
52
|
Ramos Gomes F, Romaniello V, Sánchez A, Weber C, Narayanan P, Psol M, Pardo LA. Alternatively Spliced Isoforms of KV10.1 Potassium Channels Modulate Channel Properties and Can Activate Cyclin-dependent Kinase in Xenopus Oocytes. J Biol Chem 2015; 290:30351-65. [PMID: 26518875 PMCID: PMC4683259 DOI: 10.1074/jbc.m115.668749] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 12/18/2022] Open
Abstract
KV10.1 is a voltage-gated potassium channel expressed selectively in the mammalian brain but also aberrantly in cancer cells. In this study we identified short splice variants of KV10.1 resulting from exon-skipping events (E65 and E70) in human brain and cancer cell lines. The presence of the variants was confirmed by Northern blot and RNase protection assays. Both variants completely lacked the transmembrane domains of the channel and produced cytoplasmic proteins without channel function. In a reconstituted system, both variants co-precipitated with the full-length channel and induced a robust down-regulation of KV10.1 current when co-expressed with the full-length form, but their effect was mechanistically different. E65 required a tetramerization domain and induced a reduction in the overall expression of full-length KV10.1, whereas E70 mainly affected its glycosylation pattern. E65 triggered the activation of cyclin-dependent kinases in Xenopus laevis oocytes, suggesting a role in cell cycle control. Our observations highlight the relevance of noncanonical functions for the oncogenicity of KV10.1, which need to be considered when ion channels are targeted for cancer therapy.
Collapse
Affiliation(s)
| | - Vincenzo Romaniello
- the Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Araceli Sánchez
- the Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Claudia Weber
- From the Department of Molecular Biology of Neuronal Signals and
| | | | - Maryna Psol
- From the Department of Molecular Biology of Neuronal Signals and
| | - Luis A Pardo
- the Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| |
Collapse
|
53
|
EAG2 potassium channel with evolutionarily conserved function as a brain tumor target. Nat Neurosci 2015; 18:1236-46. [PMID: 26258683 DOI: 10.1038/nn.4088] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/15/2015] [Indexed: 12/15/2022]
Abstract
Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.
Collapse
|
54
|
Bigelow AM, Khalifa MM, Clark JM. Imipramine for incessant ventricular arrhythmias in 2 unrelated patients with Andersen-Tawil syndrome. Heart Rhythm 2015; 12:1654-7. [DOI: 10.1016/j.hrthm.2015.03.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Indexed: 11/25/2022]
|
55
|
Rao VR, Perez-Neut M, Kaja S, Gentile S. Voltage-gated ion channels in cancer cell proliferation. Cancers (Basel) 2015; 7:849-75. [PMID: 26010603 PMCID: PMC4491688 DOI: 10.3390/cancers7020813] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/12/2015] [Indexed: 12/22/2022] Open
Abstract
Changes of the electrical charges across the surface cell membrane are absolutely necessary to maintain cellular homeostasis in physiological as well as in pathological conditions. The opening of ion channels alter the charge distribution across the surface membrane as they allow the diffusion of ions such as K+, Ca++, Cl.
Collapse
Affiliation(s)
- Vidhya R Rao
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago 2160 S. 1s tAve, Maywood, IL 60153, USA.
| | - Mathew Perez-Neut
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago 2160 S. 1s tAve, Maywood, IL 60153, USA.
| | - Simon Kaja
- Department of Ophthalmology and Vision Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes St., Kansas City, MO 64108, USA.
| | - Saverio Gentile
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago 2160 S. 1s tAve, Maywood, IL 60153, USA.
| |
Collapse
|
56
|
Kale VP, Amin SG, Pandey MK. Targeting ion channels for cancer therapy by repurposing the approved drugs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2747-55. [PMID: 25843679 DOI: 10.1016/j.bbamem.2015.03.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 03/18/2015] [Accepted: 03/27/2015] [Indexed: 12/21/2022]
Abstract
Ion channels have been shown to be involved in oncogenesis and efforts are being poured in to target the ion channels. There are many clinically approved drugs with ion channels as "off" targets. The question is, can these drugs be repurposed to inhibit ion channels for cancer treatment? Repurposing of drugs will not only save investors' money but also result in safer drugs for cancer patients. Advanced bioinformatics techniques and availability of a plethora of open access data on FDA approved drugs for various indications and omics data of large number of cancer types give a ray of hope to look for possibility of repurposing those drugs for cancer treatment. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Vijay Pralhad Kale
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Shantu G Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Manoj K Pandey
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
57
|
Lörinczi É, Gómez-Posada JC, de la Peña P, Tomczak AP, Fernández-Trillo J, Leipscher U, Stühmer W, Barros F, Pardo LA. Voltage-dependent gating of KCNH potassium channels lacking a covalent link between voltage-sensing and pore domains. Nat Commun 2015; 6:6672. [PMID: 25818916 PMCID: PMC4389246 DOI: 10.1038/ncomms7672] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/18/2015] [Indexed: 12/25/2022] Open
Abstract
Voltage-gated channels open paths for ion permeation upon changes in membrane potential, but how voltage changes are coupled to gating is not entirely understood. Two modules can be recognized in voltage-gated potassium channels, one responsible for voltage sensing (transmembrane segments S1 to S4), the other for permeation (S5 and S6). It is generally assumed that the conversion of a conformational change in the voltage sensor into channel gating occurs through the intracellular S4–S5 linker that provides physical continuity between the two regions. Using the pathophysiologically relevant KCNH family, we show that truncated proteins interrupted at, or lacking the S4–S5 linker produce voltage-gated channels in a heterologous model that recapitulate both the voltage-sensing and permeation properties of the complete protein. These observations indicate that voltage sensing by the S4 segment is transduced to the channel gate in the absence of physical continuity between the modules. The pore of voltage-gated ion channels opens in response to membrane depolarization sensed by a separate voltage-sensing domain. Here, Lörinczi et al. show that, contrary to assumptions, no physical linker is required to transmit changes from the voltage-sensing to the permeation domain of KCNH channels.
Collapse
Affiliation(s)
- Éva Lörinczi
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Juan Camilo Gómez-Posada
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Pilar de la Peña
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Edificio Santiago Gascón, Campus de El Cristo, 33006 Oviedo, Spain
| | - Adam P Tomczak
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Jorge Fernández-Trillo
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Ulrike Leipscher
- 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.,Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain, 37073 Göttingen, Germany
| | - Francisco Barros
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Edificio Santiago Gascón, Campus de El Cristo, 33006 Oviedo, Spain
| | - Luis A Pardo
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| |
Collapse
|
58
|
de Guadalupe Chávez-López M, Pérez-Carreón JI, Zuñiga-García V, Díaz-Chávez J, Herrera LA, Caro-Sánchez CH, Acuña-Macías I, Gariglio P, Hernández-Gallegos E, Chiliquinga AJ, Camacho J. Astemizole-based anticancer therapy for hepatocellular carcinoma (HCC), and Eag1 channels as potential early-stage markers of HCC. Tumour Biol 2015; 36:6149-58. [PMID: 25783527 DOI: 10.1007/s13277-015-3299-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 03/01/2015] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has very poor prognosis. Astemizole has gained great interest as a potential anticancer drug because it targets several proteins involved in cancer including the Eag1 (ether à-go-go-1) potassium channel that is overexpressed in human HCC. Eag1 channels are regulated by cancer etiological factors and have been proposed as early tumor markers. Here, we found that HepG2 and HuH-7 HCC cells displayed Eag1 messenger RNA (mRNA) and protein expression, determined by real-time RT-PCR and immunochemistry, respectively. Astemizole inhibited human HCC cell proliferation (assessed by metabolic activity assay) and induced apoptosis (studied with flow cytometry) in both cell lines. The subcellular Eag1 protein localization was modified by astemizole in the HepG2 cells. The treatment with astemizole prevented diethylnitrosamine (DEN)-induced rat HCC development in vivo (followed by studying γ-glutamyl transpeptidase (GGT) activity). The Eag1 mRNA and protein levels were increased in most DEN-treated groups but decreased after astemizole treatment. GGT activity was decreased by astemizole. The Eag1 protein was detected in cirrhotic and dysplastic rat livers. Astemizole might have clinical utility for HCC prevention and treatment, and Eag1 channels may be potential early HCC biomarkers. These data provide significant basis to include astemizole in HCC clinical trials.
Collapse
Affiliation(s)
- María de Guadalupe Chávez-López
- Department of Pharmacology, Centro de Investigación y de Estudios Avanzados del I.P.N., Avenida Instituto Politécnico Nacional 2508, Mexico City, 07360, Mexico
| | | | | | | | | | | | | | | | | | | | | |
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: 9.5] [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
|
Mortensen LS, Schmidt H, Farsi Z, Barrantes-Freer A, Rubio ME, Ufartes R, Eilers J, Sakaba T, Stühmer W, Pardo LA. KV 10.1 opposes activity-dependent increase in Ca²⁺ influx into the presynaptic terminal of the parallel fibre-Purkinje cell synapse. J Physiol 2014; 593:181-96. [PMID: 25556795 DOI: 10.1113/jphysiol.2014.281600] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Voltage-gated KV 10.1 potassium channels are widely expressed in the mammalian brain but their function remains poorly understood. We report that KV 10.1 is enriched in the presynaptic terminals and does not take part in somatic action potentials. In parallel fibre synapses in the cerebellar cortex, we find that KV 10.1 regulates Ca(2+) influx and neurotransmitter release during repetitive high-frequency activity. Our results describe the physiological role of mammalian KV 10.1 for the first time and help understand the fine-tuning of synaptic transmission. The voltage-gated potassium channel KV 10.1 (Eag1) is widely expressed in the mammalian brain, but its physiological function is not yet understood. Previous studies revealed highest expression levels in hippocampus and cerebellum and suggested a synaptic localization of the channel. The distinct activation kinetics of KV 10.1 indicate a role during repetitive activity of the cell. Here, we confirm the synaptic localization of KV 10.1 both biochemically and functionally and that the channel is sufficiently fast at physiological temperature to take part in repolarization of the action potential (AP). We studied the role of the channel in cerebellar physiology using patch clamp and two-photon Ca(2+) imaging in KV 10.1-deficient and wild-type mice. The excitability and action potential waveform recorded at granule cell somata was unchanged, while Ca(2+) influx into axonal boutons was enhanced in mutants in response to stimulation with three APs, but not after a single AP. Furthermore, mutants exhibited a frequency-dependent increase in facilitation at the parallel fibre-Purkinje cell synapse at high firing rates. We propose that KV 10.1 acts as a modulator of local AP shape specifically during high-frequency burst firing when other potassium channels suffer cumulative inactivation.
Collapse
Affiliation(s)
- Lena Sünke Mortensen
- Max-Planck-Institute of Experimental Medicine, 37075, Göttingen, Germany; International Max Planck Research School Neurosciences, 37077, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Kong X, Chen L, Jiao L, Jiang X, Lian F, Lu J, Zhu K, Du D, Liu J, Ding H, Zhang N, Shen J, Zheng M, Chen K, Liu X, Jiang H, Luo C. Astemizole arrests the proliferation of cancer cells by disrupting the EZH2-EED interaction of polycomb repressive complex 2. J Med Chem 2014; 57:9512-21. [PMID: 25369470 DOI: 10.1021/jm501230c] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Polycomb Repressive Complex 2 (PRC2) modulates the chromatin structure and transcriptional repression by trimethylation lysine 27 of histone H3 (H3K27me3), a process that necessitates the protein-protein interaction (PPI) between the catalytic subunit EZH2 and EED. Deregulated PRC2 is intimately involved in tumorigenesis and progression, making it an invaluable target for epigenetic cancer therapy. However, until now, there have been no reported small molecule compounds targeting the EZH2-EED interactions. In the present study, we identified astemizole, an FDA-approved drug, as a small molecule inhibitor of the EZH2-EED interaction of PRC2. The disruption of the EZH2-EED interaction by astemizole destabilizes the PRC2 complex and inhibits its methyltransferase activity in cancer cells. Multiple lines of evidence have demonstrated that astemizole arrests the proliferation of PRC2-driven lymphomas primarily by disabling the PRC2 complex. Our findings demonstrate the chemical tractability of the difficult PPI target by a small molecule compound, highlighting the therapeutic promise for PRC2-driven human cancers via targeted destruction of the EZH2-EED complex.
Collapse
Affiliation(s)
- Xiangqian Kong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Silencing of Ether à go-go 1 by shRNA inhibits osteosarcoma growth and cell cycle progression. Int J Mol Sci 2014; 15:5570-81. [PMID: 24694542 PMCID: PMC4013582 DOI: 10.3390/ijms15045570] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/18/2014] [Accepted: 03/20/2014] [Indexed: 11/17/2022] Open
Abstract
Recently, a member of the voltage-dependent potassium channel (Kv) family, the Ether à go-go 1 (Eag1) channel was found to be necessary for cell proliferation, cycle progression and tumorigenesis. However, the therapeutic potential of the Eag1 channel in osteosarcoma remains elusive. In the present study, a recombinant adenovirus harboring shRNA against Eag1 was constructed to silence Eag1 expression in human osteosarcoma MG-63 cells. We observed that Eag1-shRNA inhibited the proliferation and colony formation of MG-63 cells due to the induction of G1 phase arrest. Moreover, in vivo experiments showed that Eag1-shRNA inhibited osteosarcoma growth in a xenograft nude mice model. In addition, selective inhibition of Eag1 significantly decreased the expression levels of cyclin D1 and E. Taken together, our data suggest that the Eag1 channel plays a crucial role in regulating the proliferation and cell cycle of osteosarcoma cells, and represents a new and effective therapeutic target for osteosarcoma.
Collapse
|
63
|
Jiménez-Garduño AM, Mitkovski M, Alexopoulos IK, Sánchez A, Stühmer W, Pardo LA, Ortega A. KV10.1 K+-channel plasma membrane discrete domain partitioning and its functional correlation in neurons. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:921-31. [DOI: 10.1016/j.bbamem.2013.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 10/27/2013] [Accepted: 11/05/2013] [Indexed: 12/25/2022]
|
64
|
Schlichter LC, Jiang J, Wang J, Newell EW, Tsui FWL, Lam D. Regulation of hERG and hEAG channels by Src and by SHP-1 tyrosine phosphatase via an ITIM region in the cyclic nucleotide binding domain. PLoS One 2014; 9:e90024. [PMID: 24587194 PMCID: PMC3938566 DOI: 10.1371/journal.pone.0090024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/31/2014] [Indexed: 12/14/2022] Open
Abstract
Members of the EAG K+ channel superfamily (EAG/Kv10.x, ERG/Kv11.x, ELK/Kv12.x subfamilies) are expressed in many cells and tissues. In particular, two prototypes, EAG1/Kv10.1/KCNH1 and ERG1/Kv11.1/KCNH2 contribute to both normal and pathological functions. Proliferation of numerous cancer cells depends on hEAG1, and in some cases, hERG. hERG is best known for contributing to the cardiac action potential, and for numerous channel mutations that underlie ‘long-QT syndrome’. Many cells, particularly cancer cells, express Src-family tyrosine kinases and SHP tyrosine phosphatases; and an imbalance in tyrosine phosphorylation can lead to malignancies, autoimmune diseases, and inflammatory disorders. Ion channel contributions to cell functions are governed, to a large degree, by post-translational modulation, especially phosphorylation. However, almost nothing is known about roles of specific tyrosine kinases and phosphatases in regulating K+ channels in the EAG superfamily. First, we show that tyrosine kinase inhibitor, PP1, and the selective Src inhibitory peptide, Src40-58, reduce the hERG current amplitude, without altering its voltage dependence or kinetics. PP1 similarly reduces the hEAG1 current. Surprisingly, an ‘immuno-receptor tyrosine inhibitory motif’ (ITIM) is present within the cyclic nucleotide binding domain of all EAG-superfamily members, and is conserved in the human, rat and mouse sequences. When tyrosine phosphorylated, this ITIM directly bound to and activated SHP-1 tyrosine phosphatase (PTP-1C/PTPN6/HCP); the first report that a portion of an ion channel is a binding site and activator of a tyrosine phosphatase. Both hERG and hEAG1 currents were decreased by applying active recombinant SHP-1, and increased by the inhibitory substrate-trapping SHP-1 mutant. Thus, hERG and hEAG1 currents are regulated by activated SHP-1, in a manner opposite to their regulation by Src. Given the widespread distribution of these channels, Src and SHP-1, this work has broad implications in cell signaling that controls survival, proliferation, differentiation, and other ERG1 and EAG1 functions in many cell types.
Collapse
Affiliation(s)
- Lyanne C. Schlichter
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| | - Jiahua Jiang
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - John Wang
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Evan W. Newell
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology University of Toronto, Toronto, Ontario, Canada
| | - Florence W. L. Tsui
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Doris Lam
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
65
|
Zhang YY, Yue J, Che H, Sun HY, Tse HF, Li GR. BKCaand hEag1 Channels Regulate Cell Proliferation and Differentiation in Human Bone Marrow-Derived Mesenchymal Stem Cells. J Cell Physiol 2013; 229:202-12. [DOI: 10.1002/jcp.24435] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 07/15/2013] [Indexed: 12/29/2022]
Affiliation(s)
- Ying-Ying Zhang
- Department of Medicine; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
| | - Jianbo Yue
- Department of Physiology; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
| | - Hui Che
- Department of Medicine; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
| | - Hai-Ying Sun
- Department of Medicine; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
- Department of Physiology; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
| | - Hung-Fat Tse
- Department of Medicine; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
| | - Gui-Rong Li
- Department of Medicine; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
- Department of Physiology; Li Ka Shing Faculty of Medicine; University of Hong Kong; Hong Kong China
| |
Collapse
|
66
|
Zhao F. Flow Injection Post-Chemiluminescence Reaction of Astemizole in N-Bromosuccinimide-Calcein System and Its Application. ANAL LETT 2013. [DOI: 10.1080/00032719.2012.689904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
67
|
Oshima A, Hirano-Iwata A, Mozumi H, Ishinari Y, Kimura Y, Niwano M. Reconstitution of Human Ether-a-go-go-Related Gene Channels in Microfabricated Silicon Chips. Anal Chem 2013; 85:4363-9. [DOI: 10.1021/ac303484k] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Azusa Oshima
- Graduate School of Biomedical
Engineering, Tohoku University, 6-6 Aoba,
Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Ayumi Hirano-Iwata
- Graduate School of Biomedical
Engineering, Tohoku University, 6-6 Aoba,
Aramaki, Aoba-ku, Sendai 980-8579, Japan
- PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi,
Saitama 332-0012, Japan
| | - Hideki Mozumi
- Graduate School of Biomedical
Engineering, Tohoku University, 6-6 Aoba,
Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Yutaka Ishinari
- Graduate School of Biomedical
Engineering, Tohoku University, 6-6 Aoba,
Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Yasuo Kimura
- Laboratory
for Nanoelectronics
and Spintronics, Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai,
Miyagi 980-8577, Japan
| | - Michio Niwano
- Graduate School of Biomedical
Engineering, Tohoku University, 6-6 Aoba,
Aramaki, Aoba-ku, Sendai 980-8579, Japan
- Laboratory
for Nanoelectronics
and Spintronics, Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai,
Miyagi 980-8577, Japan
| |
Collapse
|
68
|
Ufartes R, Schneider T, Mortensen LS, de Juan Romero C, Hentrich K, Knoetgen H, Beilinson V, Moebius W, Tarabykin V, Alves F, Pardo LA, Rawlins JNP, Stuehmer W. Behavioural and functional characterization of Kv10.1 (Eag1) knockout mice. Hum Mol Genet 2013; 22:2247-62. [PMID: 23424202 PMCID: PMC3652421 DOI: 10.1093/hmg/ddt076] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Kv10.1 (Eag1), member of the Kv10 family of voltage-gated potassium channels, is preferentially expressed in adult brain. The aim of the present study was to unravel the functional role of Kv10.1 in the brain by generating knockout mice, where the voltage sensor and pore region of Kv10.1 were removed to render non-functional proteins through deletion of exon 7 of the KCNH1 gene using the ‘3 Lox P strategy’. Kv10.1-deficient mice show no obvious alterations during embryogenesis and develop normally to adulthood; cortex, hippocampus and cerebellum appear anatomically normal. Other tests, including general health screen, sensorimotor functioning and gating, anxiety, social behaviour, learning and memory did not show any functional aberrations in Kv10.1 null mice. Kv10.1 null mice display mild hyperactivity and longer-lasting haloperidol-induced catalepsy, but there was no difference between genotypes in amphetamine sensitization and withdrawal, reactivity to apomorphine and haloperidol in the prepulse inhibition tests or to antidepressants in the haloperidol-induced catalepsy. Furthermore, electrical properties of Kv10.1 in cerebellar Purkinje cells did not show any difference between genotypes. Bearing in mind that Kv10.1 is overexpressed in over 70% of all human tumours and that its inhibition leads to a reduced tumour cell proliferation, the fact that deletion of Kv10.1 does not show a marked phenotype is a prerequisite for utilizing Kv10.1 blocking and/or reduction techniques, such as siRNA, to treat cancer.
Collapse
Affiliation(s)
- Roser Ufartes
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, Göttingen 37077, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
p38 MAPK regulates the expression of ether à go-go potassium channel in human osteosarcoma cells. Radiol Oncol 2013; 47:42-9. [PMID: 23450231 PMCID: PMC3573833 DOI: 10.2478/v10019-012-0043-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 05/30/2012] [Indexed: 12/14/2022] Open
Abstract
Background The ether à go-go (Eag) channel has been shown to be overexpressed in a variety of cancers. However, the expression and function of Eag in osteosarcoma are poorly understood. In addition, the molecular mechanisms responsible for Eag overexpression in cancer cells remain unclear. Methods The expression of Eag in human osteosarcoma cell line MG-63 was detected by reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis. The effect of Eag inhibition on MG-63 cell proliferation was assessed in vitro. The effect of short hairpin RNA (shRNA) mediated knockdown of Eag on osteosarcoma growth was evaluated in xenograft model in vivo. The activation of mitogen-activated protein kinase (MAPK) pathway and p53 in MG-63 cells was detected by Western blot analysis. Results Eag was overexpressed in MG-63 cells. Imipramine or Eag shRNA significantly suppressed the proliferation of MG-63 cells in vitro and in vivo. MG-63 cell proliferation was specifically inhibited by p38 MAPK inhibitor SB203580 or small interference RNA (siRNA). The inhibition of p38 MAPK activation by SB203580 or siRNA reduced Eag protein level but increased p53 protein level. Moreover, the activation of p53 by nutlin-3 induced cell growth arrest in MG-63 cells and reduced Eag protein level, while the inactivation of p53 by pifithrin-alpha (PFT-α) promoted MG-63 cell growth and increased Eag protein expression. Conclusions Eag channel functions as an oncogene to promote the proliferation of human osteosarocma cells. Furthermore, the high expression of Eag in osteosarcoma cells is regulated by p38 MAPK/p53 pathway.
Collapse
|
70
|
Wu X, Zhong D, Gao Q, Zhai W, Ding Z, Wu J. MicroRNA-34a inhibits human osteosarcoma proliferation by downregulating ether à go-go 1 expression. Int J Med Sci 2013; 10:676-82. [PMID: 23569431 PMCID: PMC3619116 DOI: 10.7150/ijms.5528] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 03/21/2013] [Indexed: 12/15/2022] Open
Abstract
Aberrant expression of MicroRNAs (miRNAs) has been implicated in several types of cancer. As a direct target gene of p53, miR-34a has been suggested to mediate the tumor suppressor function of p53. Ether à go-go 1 (Eag1) channel is overexpressed in a variety of cancers and plays important roles in cancer progression. However, the link between miR-34a and Eag1 in cancer is unclear. In this study, we used human osteosarcoma as the model to demonstrate that miR-34a was significantly downregulated in osteosarcoma tissues and cell lines compared with normal brain tissues and osteoblastic cell line. Next we evaluated the role of miR-34a in the regulation of osteosarcoma cell proliferation by CCK-8 and colony formation assays. The results showed that overexpression of miR-34a inhibited the proliferation of MG-63 and Saos-2 cells. Furthermore, xenograft nude mice model showed that miR-34a inhibited osteosarcoma growth in vivo. Mechanistically, we found that overexpression of miR-34a led to decreased Eag1 expression in osteosarcoma cells while inhibition of miR-34a increased Eag1 expression. Taken together, our results suggest that miR-34a could inhibit osteosarcoma growth via the down regulation of Eag1 expression.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Neurology, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou 363000, China
| | | | | | | | | | | |
Collapse
|
71
|
Garg V, Sachse FB, Sanguinetti MC. Tuning of EAG K(+) channel inactivation: molecular determinants of amplification by mutations and a small molecule. ACTA ACUST UNITED AC 2012; 140:307-24. [PMID: 22930803 PMCID: PMC3434097 DOI: 10.1085/jgp.201210826] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Ether-à-go-go (EAG) and EAG-related gene (ERG) K+ channels are close homologues but differ markedly in their gating properties. ERG1 channels are characterized by rapid and extensive C-type inactivation, whereas mammalian EAG1 channels were previously considered noninactivating. Here, we show that human EAG1 channels exhibit an intrinsic voltage-dependent slow inactivation that is markedly enhanced in rate and extent by 1–10 µM 3-nitro-N-(4-phenoxyphenyl) benzamide, or ICA105574 (ICA). This compound was previously reported to have the opposite effect on ERG1 channels, causing an increase in current magnitude by inhibition of C-type inactivation. The voltage dependence of 2 µM ICA-induced inhibition of EAG1 current was half-maximal at −73 mV, 62 mV negative to the half-point for channel activation. This finding suggests that current inhibition by the drug is mediated by enhanced inactivation and not open-channel block, where the voltage half-points for current inhibition and channel activation are predicted to overlap, as we demonstrate for clofilium and astemizole. The mutation Y464A in the S6 segment also induced inactivation of EAG1, with a time course and voltage dependence similar to that caused by 2 µM ICA. Several Markov models were investigated to describe gating effects induced by multiple concentrations of the drug and the Y464A mutation. Models with the smallest fit error required both closed- and open-state inactivation. Unlike typical C-type inactivation, the rate of Y464A- and ICA-induced inactivation was not decreased by external tetraethylammonium or elevated [K+]e. EAG1 channel inactivation introduced by Y464A was prevented by additional mutation of a nearby residue located in the S5 segment (F359A) or pore helix (L434A), suggesting a tripartite molecular model where interactions between single residues in S5, S6, and the pore helix modulate inactivation of EAG1 channels.
Collapse
Affiliation(s)
- Vivek Garg
- Department of Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
72
|
Mobasheri A, Lewis R, Ferreira-Mendes A, Rufino A, Dart C, Barrett-Jolley R. Potassium channels in articular chondrocytes. Channels (Austin) 2012; 6:416-25. [PMID: 23064164 PMCID: PMC3536726 DOI: 10.4161/chan.22340] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chondrocytes are the resident cells of cartilage, which synthesize and maintain the extracellular matrix. The range of known potassium channels expressed by these unique cells is continually increasing. Since chondrocytes are non-excitable, and do not need to be repolarized following action potentials, the function of potassium channels in these cells has, until recently, remained completely unknown. However, recent advances in both traditional physiology and “omic” technologies have enhanced our knowledge and understanding of the chondrocyte channelome. A large number of potassium channels have been identified and a number of putative, but credible, functions have been proposed. Members of each of the potassium channel sub-families (calcium activated, inward rectifier, voltage-gated and tandem pore) have all been identified. Mechanotransduction, cell volume regulation, apoptosis and chondrogenesis all appear to involve potassium channels. Since evidence suggests that potassium channel gene transcription is altered in osteoarthritis, future studies are needed that investigate potassium channels as potential cellular biomarkers and therapeutic targets for treatment of degenerative joint conditions.
Collapse
Affiliation(s)
- Ali Mobasheri
- Musculoskeletal Research Group, Division of Veterinary Medicine, Faculty of Medicine and Health Sciences, The University of Nottingham, Sutton Bonington Campus, Sutton Bonington, Leicestershire, UK. ali.
| | | | | | | | | | | |
Collapse
|
73
|
García-Quiroz J, García-Becerra R, Barrera D, Santos N, Avila E, Ordaz-Rosado D, Rivas-Suárez M, Halhali A, Rodríguez P, Gamboa-Domínguez A, Medina-Franco H, Camacho J, Larrea F, Díaz L. Astemizole synergizes calcitriol antiproliferative activity by inhibiting CYP24A1 and upregulating VDR: a novel approach for breast cancer therapy. PLoS One 2012; 7:e45063. [PMID: 22984610 PMCID: PMC3440370 DOI: 10.1371/journal.pone.0045063] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 08/14/2012] [Indexed: 11/23/2022] Open
Abstract
Background Calcitriol antiproliferative effects include inhibition of the oncogenic ether-à-go-go-1 potassium channel (Eag1) expression, which is necessary for cell cycle progression and tumorigenesis. Astemizole, a new promising antineoplastic drug, targets Eag1 by blocking ion currents. Herein, we characterized the interaction between calcitriol and astemizole as well as their conjoint antiproliferative action in SUM-229PE, T-47D and primary tumor-derived breast cancer cells. Methodology/Principal Findings Molecular markers were studied by immunocytochemistry, Western blot and real time PCR. Inhibitory concentrations were determined by dose-response curves and metabolic activity assays. At clinically achievable drug concentrations, synergistic antiproliferative interaction was observed between calcitriol and astemizole, as calculated by combination index analysis (CI <1). Astemizole significantly enhanced calcitriol’s growth-inhibitory effects (3–11 folds, P<0.01). Mean IC20 values were 1.82±2.41 nM and 1.62±0.75 µM; for calcitriol (in estrogen receptor negative cells) and astemizole, respectively. Real time PCR showed that both drugs alone downregulated, while simultaneous treatment further reduced Ki-67 and Eag1 gene expression (P<0.05). Astemizole inhibited basal and calcitriol-induced CYP24A1 and CYP3A4 mRNA expression (cytochromes involved in calcitriol and astemizole degradation) in breast and hepatoma cancer cells, respectively, while upregulated vitamin D receptor (VDR) expression. Conclusions/Significance Astemizole synergized calcitriol antiproliferative effects by downregulating CYP24A1, upregulating VDR and targeting Eag1. This study provides insight into the molecular mechanisms involved in astemizole-calcitriol combined antineoplastic effect, offering scientific support to test both compounds in combination in further preclinical and clinical studies of neoplasms expressing VDR and Eag1. VDR-negative tumors might also be sensitized to calcitriol antineoplastic effects by the use of astemizole. Herein we suggest a novel combined adjuvant therapy for the management of VDR/Eag1-expressing breast cancer tumors. Since astemizole improves calcitriol bioavailability and activity, decreased calcitriol dosing is advised for conjoint administration.
Collapse
Affiliation(s)
- Janice García-Quiroz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N., México, D.F., México
| | - Rocío García-Becerra
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - David Barrera
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Nancy Santos
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N., México, D.F., México
| | - Euclides Avila
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - David Ordaz-Rosado
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Mariana Rivas-Suárez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Ali Halhali
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Pamela Rodríguez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Armando Gamboa-Domínguez
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Heriberto Medina-Franco
- Departamento de Cirugía, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N., México, D.F., México
| | - Fernando Larrea
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, D.F., México
- * E-mail:
| |
Collapse
|
74
|
Zhang J, Chan YC, Ho JCY, Siu CW, Lian Q, Tse HF. Regulation of cell proliferation of human induced pluripotent stem cell-derived mesenchymal stem cells via ether-à-go-go 1 (hEAG1) potassium channel. Am J Physiol Cell Physiol 2012; 303:C115-25. [DOI: 10.1152/ajpcell.00326.2011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The successful generation of a high yield of mesenchymal stem cells (MSCs) from human induced pluripotent stem cells (iPSCs) may represent an unlimited cell source with superior therapeutic benefits for tissue regeneration to bone marrow (BM)-derived MSCs. We investigated whether the differential expression of ion channels in iPSC-MSCs was responsible for their higher proliferation capacity than BM-MSCs. The expression of ion channels for K+, Na+, Ca2+, and Cl− was examined by RT-PCR. The electrophysiological properties of iPSC-MSCs and BM-MSCs were then compared by patch-clamp experiments to verify their functional roles. Significant mRNA expression of ion channel genes including KCa1.1, KCa3.1, KCNH1, Kir2.1, SCN9A, CACNA1C, and Clcn3 was observed in both human iPSC-MSCs and BM-MSCs, whereas Kir2.2 and Kir2.3 were only detected in human iPSC-MSCs. Five types of currents [big-conductance Ca2+-activated K+ current (BKCa), delayed rectifier K+ current ( IKDR), inwardly rectifying K+ current ( IKir), Ca2+-activated K+ current ( IKCa), and chloride current ( ICl)] were found in iPSC-MSCs (83%, 47%, 11%, 5%, and 4%, respectively) but only four of them (BKCa, IKDR, IKir, and IKCa) were identified in BM-MSCs (76%, 25%, 22%, and 11%, respectively). Cell proliferation was examined with MTT or bromodeoxyuridine assay, and doubling times were 2.66 and 3.72 days for iPSC-MSCs and BM-MSCs, respectively, showing a 1.4-fold discrepancy. Blockade of IKDR with short hairpin RNA or human ether-à-go-go 1 (hEAG1) channel blockers, 4-AP and astemizole, significantly reduced the rate of proliferation of human iPSC-MSCs. These treatments also decreased the rate of proliferation of human BM-MSCs albeit to a lesser extent. These findings demonstrate that the hEAG1 channel plays a crucial role in controlling the proliferation rate of human iPSC-MSCs and to a lesser extent in BM-MSCs.
Collapse
Affiliation(s)
- Jiao Zhang
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
| | - Yau-Chi Chan
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
| | - Jenny Chung-Yee Ho
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
- Research Centre of Heart, Brain, Hormone, and Healthy Aging, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong; and
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
- Research Centre of Heart, Brain, Hormone, and Healthy Aging, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong; and
| | - Qizhou Lian
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
- Research Centre of Heart, Brain, Hormone, and Healthy Aging, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong; and
- Eye Institute, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
- Research Centre of Heart, Brain, Hormone, and Healthy Aging, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong; and
| |
Collapse
|
75
|
Dennis AT, Wang L, Wan H, Nassal D, Deschenes I, Ficker E. Molecular determinants of pentamidine-induced hERG trafficking inhibition. Mol Pharmacol 2011; 81:198-209. [PMID: 22046004 DOI: 10.1124/mol.111.075135] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pentamidine is an antiprotozoal compound that clinically causes acquired long QT syndrome (acLQTS), which is associated with prolonged QT intervals, tachycardias, and sudden cardiac arrest. Pentamidine delays terminal repolarization in human heart by acutely blocking cardiac inward rectifier currents. At the same time, pentamidine reduces surface expression of the cardiac potassium channel I(Kr)/human ether à-go-go-related gene (hERG). This is unusual in that acLQTS is caused most often by direct block of the cardiac potassium current I(Kr)/hERG. The present study was designed to provide a more complete picture of how hERG surface expression is disrupted by pentamidine at the cellular and molecular levels. Using biochemical and electrophysiological methods, we found that pentamidine exclusively inhibits hERG export from the endoplasmic reticulum to the cell surface in a heterologous expression system as well as in cardiomyocytes. hERG trafficking inhibition could be rescued in the presence of the pharmacological chaperone astemizole. We used rescue experiments in combination with an extensive mutational analysis to locate an interaction site for pentamidine at phenylalanine 656, a crucial residue in the canonical drug binding site of terminally folded hERG. Our data suggest that pentamidine binding to a folding intermediate of hERG arrests channel maturation in a conformational state that cannot be exported from the endoplasmic reticulum. We propose that pentamidine is the founding member of a novel pharmacological entity whose members act as small molecule antichaperones.
Collapse
Affiliation(s)
- Adrienne T Dennis
- Rammelkamp Center for Education and Research, MetroHealth Campus, Cleveland, OH 44109, USA
| | | | | | | | | | | |
Collapse
|
76
|
Kohl T, Lörinczi E, Pardo LA, Stühmer W. Rapid internalization of the oncogenic K+ channel K(V)10.1. PLoS One 2011; 6:e26329. [PMID: 22022602 PMCID: PMC3192180 DOI: 10.1371/journal.pone.0026329] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/24/2011] [Indexed: 11/18/2022] Open
Abstract
K(V)10.1 is a mammalian brain voltage-gated potassium channel whose ectopic expression outside of the brain has been proven relevant for tumor biology. Promotion of cancer cell proliferation by K(V)10.1 depends largely on ion flow, but some oncogenic properties remain in the absence of ion permeation. Additionally, K(V)10.1 surface populations are small compared to large intracellular pools. Control of protein turnover within cells is key to both cellular plasticity and homeostasis, and therefore we set out to analyze how endocytic trafficking participates in controlling K(V)10.1 intracellular distribution and life cycle. To follow plasma membrane K(V)10.1 selectively, we generated a modified channel of displaying an extracellular affinity tag for surface labeling by α-bungarotoxin. This modification only minimally affected K(V)10.1 electrophysiological properties. Using a combination of microscopy and biochemistry techniques, we show that K(V)10.1 is constitutively internalized involving at least two distinct pathways of endocytosis and mainly sorted to lysosomes. This occurs at a relatively fast rate. Simultaneously, recycling seems to contribute to maintain basal K(V)10.1 surface levels. Brief K(V)10.1 surface half-life and rapid lysosomal targeting is a relevant factor to be taken into account for potential drug delivery and targeting strategies directed against K(V)10.1 on tumor cells.
Collapse
Affiliation(s)
- Tobias Kohl
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Biology of Neuronal Signals, Göttingen, Germany
| | - Eva Lörinczi
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Biology of Neuronal Signals, Göttingen, Germany
| | - Luis A. Pardo
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Biology of Neuronal Signals, Göttingen, Germany
| | - Walter Stühmer
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Biology of Neuronal Signals, Göttingen, Germany
- DFG Research Center for Molecular Physiology of the Brain (CMPB), Göttingen, Germany
| |
Collapse
|
77
|
Tumor cell-selective apoptosis induction through targeting of K(V)10.1 via bifunctional TRAIL antibody. Mol Cancer 2011; 10:109. [PMID: 21899742 PMCID: PMC3179451 DOI: 10.1186/1476-4598-10-109] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 09/07/2011] [Indexed: 11/26/2022] Open
Abstract
Background The search for strategies to target ion channels for therapeutic applications has become of increasing interest. Especially, the potassium channel KV10.1 (Ether-á-go-go) is attractive as target since this surface protein is virtually not detected in normal tissue outside the central nervous system, but is expressed in approximately 70% of tumors from different origins. Methods We designed a single-chain antibody against an extracellular region of KV10.1 (scFv62) and fused it to the human soluble TRAIL. The KV10.1-specific scFv62 antibody -TRAIL fusion protein was expressed in CHO-K1 cells, purified by chromatography and tested for biological activity. Results Prostate cancer cells, either positive or negative for KV10.1 were treated with the purified construct. After sensitization with cytotoxic drugs, scFv62-TRAIL induced apoptosis only in KV10.1-positive cancer cells, but not in non-tumor cells, nor in tumor cells lacking KV10.1 expression. In co-cultures with KV10.1-positive cancer cells the fusion protein also induced apoptosis in bystander KV10.1-negative cancer cells, while normal prostate epithelial cells were not affected when present as bystander. Conclusions KV10.1 represents a novel therapeutic target for cancer. We could design a strategy that selectively kills tumor cells based on a KV10.1-specific antibody.
Collapse
|
78
|
Wallace JL, Gow IF, Warnock M. The life and death of breast cancer cells: proposing a role for the effects of phytoestrogens on potassium channels. J Membr Biol 2011; 242:53-67. [PMID: 21728044 DOI: 10.1007/s00232-011-9376-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 06/20/2011] [Indexed: 10/18/2022]
Abstract
Changes in the regulation of potassium channels are increasingly implicated in the altered activity of breast cancer cells. Increased or reduced expression of a number of K(+) channels have been identified in numerous breast cancer cell lines and cancerous tissue biopsy samples, compared to normal tissue, and are associated with tumor formation and spread, enhanced levels of proliferation, and resistance to apoptotic stimuli. Through knockout or silencing of K(+) channel genes, and use of specific or more broad pharmacologic K(+) channel blockers, the growth of numerous cell lines, including breast cancer cells, has been modified. In this manner it has been proposed that in MCF7 breast cancer cells proliferation appears to be regulated by the activity of a number of K(+) channels, including the Ca(2+) activated K(+) channels, and the voltage-gated K(+) channels hEAG and K(v)1.1. The effect of phytoestrogens on K(+) channels has not been extensively studied but yields some interesting results. In a number of cell lines the phytoestrogen genistein inhibits K(+) current through several channels including K(v)1.3 and hERG. Where it has been used, structurally similar daidzein has little or no effect on K(+) channel activity. Since many K(+) channels have roles in proliferation and apoptosis in breast cancer cells, the impact of K(+) channel regulation by phytoestrogens is of potentially great relevance.
Collapse
Affiliation(s)
- Joanne L Wallace
- School of Health Sciences, Queen Margaret University, Musselburgh, Edinburgh, Scotland, UK.
| | | | | |
Collapse
|
79
|
Li M, Xiong ZG. Ion channels as targets for cancer therapy. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2011; 3:156-166. [PMID: 21760973 PMCID: PMC3134009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 06/26/2011] [Indexed: 05/31/2023]
Abstract
Cancer is a leading cause of death in the world. Conventional treatments have severe side effects and low survival rate. It is important to discover new targets and therapeutic strategies to improve the clinical outcomes of cancer patients. Ion channels are specialized membrane proteins that play important roles in various physiological processes. Recent studies have shown that abnormal expression and/or activity of a number of ion channels e.g. voltage-gated K(+), Na(+), Ca(2+) channels, TRP channels, and epithelial Na(+)/degenerin family of ion channels, are involved in the growth/proliferation, migration and/or invasion of cancer cells. In this review, we summarize the present knowledge about the roles of different ion channels in the development of cancer.
Collapse
Affiliation(s)
- Minghua Li
- Department of Psychology, Washington State University, VancouverWA, USA
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of MedicineAtlanta, GA, USA
| |
Collapse
|
80
|
Borowiec AS, Hague F, Gouilleux-Gruart V, Lassoued K, Ouadid-Ahidouch H. Regulation of IGF-1-dependent cyclin D1 and E expression by hEag1 channels in MCF-7 cells: The critical role of hEag1 channels in G1 phase progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:723-30. [DOI: 10.1016/j.bbamcr.2011.01.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 01/19/2011] [Accepted: 01/24/2011] [Indexed: 11/30/2022]
|
81
|
Asher V, Warren A, Shaw R, Sowter H, Bali A, Khan R. The role of Eag and HERG channels in cell proliferation and apoptotic cell death in SK-OV-3 ovarian cancer cell line. Cancer Cell Int 2011; 11:6. [PMID: 21392380 PMCID: PMC3063814 DOI: 10.1186/1475-2867-11-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 03/10/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The voltage gated potassium (K+) channels Eag and HERG have been implicated in the pathogenesis of various cancers, through association with cell cycle changes and programmed cell death. The role of these channels in the onset and progression of ovarian cancer is unknown. An understanding of mechanism by which Eag and HERG channels affect cell proliferation in ovarian cancer cells is required and therefore we investigated their role in cell proliferation and their effect on the cell cycle and apoptosis of ovarian cancer cells. METHODS The presence of Eag and HERG was determined in SK-OV-3 cells using immunofluorescence and western blotting. The effect of the Eag blockers (imipramine and clofilium) and HERG blockers (E-4031 and ergtoxin) on cell proliferation was assessed using the MTS assay with further investigation of their role in the cell cycle and apoptosis determined by flow cytometry. RESULTS Eag and HERG channels were present in the cytoplasm and nuclei of SK-OV-3 cells. There was significant inhibition of proliferation of SK-OV-3 cells by imipramine (P < 0.001) and ergtoxin (P < 0.05) at 72 hours of culture. Incubation of cells with ergtoxin led to the accumulation of cells in the S and G2/M phase, while cells accumulated in S phase after incubation with E-4031, with no effect on apoptosis. Imipramine did not affect the cell cycle but increased the proportion of SK-OV-3 cells undergoing early apoptosis. CONCLUSION Both Eag and HERG channels are expressed in SK-OV-3 ovarian cancer cells and have a role in cell proliferation. HERG channels affect the cell cycle while Eag channels are implicated in the inhibition of apoptosis of ovarian cancer cells. The family of Eag channels may represent a new therapeutic target for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Viren Asher
- School of Graduate Entry Medicine and Health Royal Derby Hospital, Uttoxeter Road, Derby DE22 3DT, UK.
| | | | | | | | | | | |
Collapse
|
82
|
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: 56] [Impact Index Per Article: 4.0] [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
|
83
|
Martin S, Lino-de-Oliveira C, Joca SRL, Weffort de Oliveira R, Echeverry MB, Da Silva CA, Pardo L, Stühmer W, Bel ED. Eag 1, Eag 2 and Kcnn3 gene brain expression of isolated reared rats. GENES BRAIN AND BEHAVIOR 2010; 9:918-24. [DOI: 10.1111/j.1601-183x.2010.00632.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
84
|
Castle NA. Pharmacological modulation of voltage-gated potassium channels as a therapeutic strategy. Expert Opin Ther Pat 2010; 20:1471-503. [PMID: 20726689 DOI: 10.1517/13543776.2010.513384] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE OF THE FIELD The human genome encodes at least 40 distinct voltage-gated potassium channel subtypes, which vary in regional expression, pharmacological and biophysical properties. Voltage-dependent potassium (Kv) channels help orchestrate many of the physiological and pathophysiological processes that promote and sometimes hinder the healthy functioning of our bodies. AREAS COVERED IN THIS REVIEW This review summarizes patent and scientific literature reports from the past decade highlighting the opportunities that Kv channels offer for the development of new therapeutic interventions for a wide variety of disorders. WHAT THE READER WILL GAIN The reader will gain an insight from an analysis of the associations of different Kv family members with disease processes, summary and evaluation of the development of therapeutically relevant pharmacological modulators of these channels, particularly focusing on proprietary agents being developed. TAKE HOME MESSAGE Development of new drugs that target Kv channels continue to be of great interest but is proving to be challenging. Nevertheless, opportunities for Kv channel modulators to have an impact on a wide range of disorders in the future remain an exciting prospect.
Collapse
|
85
|
Lee EH, Oh JH, Park HJ, Kim DG, Lee JH, Kim CY, Kwon MS, Yoon S. Simultaneous gene expression signature of heart and peripheral blood mononuclear cells in astemizole-treated rats. Arch Toxicol 2010; 84:609-18. [DOI: 10.1007/s00204-010-0529-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 02/22/2010] [Indexed: 01/10/2023]
|
86
|
Agarwal JR, Griesinger F, Stühmer W, Pardo LA. The potassium channel Ether à go-go is a novel prognostic factor with functional relevance in acute myeloid leukemia. Mol Cancer 2010; 9:18. [PMID: 20105281 PMCID: PMC2835655 DOI: 10.1186/1476-4598-9-18] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 01/27/2010] [Indexed: 11/16/2022] Open
Abstract
Background The voltage-gated potassium channel hEag1 (KV10.1) has been related to cancer biology. The physiological expression of the human channel is restricted to the brain but it is frequently and abundantly expressed in many solid tumors, thereby making it a promising target for a specific diagnosis and therapy. Because chronic lymphatic leukemia has been described not to express hEag1, it has been assumed that the channel is not expressed in hematopoietic neoplasms in general. Results Here we show that this assumption is not correct, because the channel is up-regulated in myelodysplastic syndromes, chronic myeloid leukemia and almost half of the tested acute myeloid leukemias in a subtype-dependent fashion. Most interestingly, channel expression strongly correlated with increasing age, higher relapse rates and a significantly shorter overall survival. Multivariate Cox regression analysis revealed hEag1 expression levels in AML as an independent predictive factor for reduced disease-free and overall survival; such an association had not been reported before. As a functional correlate, specific hEag1 blockade inhibited the proliferation and migration of several AML cell lines and primary cultured AML cells in vitro. Conclusion Our observations implicate hEag1 as novel target for diagnostic, prognostic and/or therapeutic approaches in AML.
Collapse
Affiliation(s)
- Jasmin R Agarwal
- Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str, 3, 37075 Göttingen, Germany
| | | | | | | |
Collapse
|
87
|
Kato S, Kato Y, Nakamura T, Sugiura T, Kubo Y, Deguchi Y, Tsuji A. Genetic deficiency of carnitine/organic cation transporter 2 (slc22a5) is associated with altered tissue distribution of its substrate pyrilamine in mice. Biopharm Drug Dispos 2010; 30:495-507. [PMID: 19821448 DOI: 10.1002/bdd.681] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Carnitine/organic cation transporter 2 (OCTN2) recognizes various cationic compounds as substrates in vitro, but information on its pharmacokinetic role in vivo is quite limited. This paper demonstrates altered tissue distribution of the OCTN2 substrate pyrilamine in juvenile visceral steatosis (jvs) mice, which have a hereditary defect of the octn2 gene. At 30 min after intravenous injection of pyrilamine, the tissue-to-plasma concentration ratio (K(p)) in the heart and pancreas was higher, whereas the K(p) in kidney and testis was lower in jvs mice compared with wild-type mice. Pyrilamine transport studies in isolated heart slices confirmed higher accumulation, together with lower efflux, of pyrilamine in the heart of jvs mice. The higher accumulation in heart slices of jvs mice was abolished by lowering the temperature, by increasing the substrate concentration, and in the presence of other H(1) antagonists or another OCTN2 substrate, carnitine, suggesting that OCTN2 extrudes pyrilamine from heart tissue. On the other hand, the lower distribution to the kidney of jvs mice was probably due to down-regulation of a basolateral transporter coupled with OCTN2, because, in jvs mice, (i) the K(p) of pyrilamine in kidney assessed immediately after intravenous injection (approximately 1 min) was also lower, (ii) the urinary excretion of pyrilamine was lower, and (iii) the uptake of pyrilamine in kidney slices was lower. The renal uptake of pyrilamine was saturable (K(m) approximately 236 microM) and was strongly inhibited by cyproheptadine, astemizole, ebastine and terfenadine. The present study thus indicates that genetic deficiency of octn2 alters pyrilamine disposition tissue-dependently.
Collapse
Affiliation(s)
- Sayaka Kato
- Division of Pharmaceutical Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | | | |
Collapse
|
88
|
Díaz L, Ceja-Ochoa I, Restrepo-Angulo I, Larrea F, Avila-Chávez E, García-Becerra R, Borja-Cacho E, Barrera D, Ahumada E, Gariglio P, Alvarez-Rios E, Ocadiz-Delgado R, Garcia-Villa E, Hernández-Gallegos E, Camacho-Arroyo I, Morales A, Ordaz-Rosado D, García-Latorre E, Escamilla J, Sánchez-Peña LC, Saqui-Salces M, Gamboa-Dominguez A, Vera E, Uribe-Ramírez M, Murbartián J, Ortiz CS, Rivera-Guevara C, De Vizcaya-Ruiz A, Camacho J. Estrogens and human papilloma virus oncogenes regulate human ether-à-go-go-1 potassium channel expression. Cancer Res 2009; 69:3300-7. [PMID: 19351862 DOI: 10.1158/0008-5472.can-08-2036] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ether-à-go-go-1 (Eag1) potassium channels are potential tools for detection and therapy of numerous cancers. Here, we show human Eag1 (hEag1) regulation by cancer-associated factors. We studied hEag1 gene expression and its regulation by estradiol, antiestrogens, and human papillomavirus (HPV) oncogenes (E6/E7). Primary cultures from normal placentas and cervical cancer tissues; tumor cell lines from cervix, choriocarcinoma, keratinocytes, and lung; and normal cell lines from vascular endothelium, keratinocytes, and lung were used. Reverse transcription-PCR (RT-PCR) experiments and Southern blot analysis showed Eag1 expression in all of the cancer cell types, normal trophoblasts, and vascular endothelium, in contrast to normal keratinocytes and lung cells. Estradiol and antiestrogens regulated Eag1 in a cell type-dependent manner. Real-time RT-PCR experiments in HeLa cells showed that Eag1 estrogenic regulation was strongly associated with the expression of estrogen receptor-alpha. Eag1 protein was detected by monoclonal antibodies in normal placenta and placental blood vessels. Patch-clamp recordings in normal trophoblasts treated with estradiol exhibited potassium currents resembling Eag1 channel activity. Eag1 gene expression in keratinocytes depended either on cellular immortalization or the presence of HPV oncogenes. Eag1 protein was found in keratinocytes transfected with E6/E7 HPV oncogenes. Cell proliferation of E6/E7 keratinocytes was decreased by Eag1 antibodies inhibiting channel activity and by the nonspecific Eag1 inhibitors imipramine and astemizole; the latter also increased apoptosis. Our results propose novel oncogenic mechanisms of estrogen/antiestrogen use and HPV infection. We also suggest Eag1 as an early indicator of cell proliferation leading to malignancies and a therapeutic target at early stages of cellular hyperproliferation.
Collapse
Affiliation(s)
- Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Ding XW, Gao S, Wang W, Yan JJ, Luo HS. Effect of imipramine on HT-29 cells' proliferation, cell cycle arrest and apoptosis and its mechanism. Shijie Huaren Xiaohua Zazhi 2008; 16:3043-3049. [DOI: 10.11569/wcjd.v16.i27.3043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of imipramine on cell growth, cell cycle and apoptosis of HT-29 colon cancer cells, and to elucidate its molecular mechanism.
METHODS: Human colon cancer HT-29 cells were grown with routine cell cultivation and cells were treated with different concentrations of imiprmine. Cell survival was determined using MTT assay at 24 h, 48 h and 72 h, respectively; cell cycle distribution was assessed by FACS flow cytometery after propidium iodide staining; apoptosis of HT-29 cells was detected using Annexin V/PI methods and DNA ladder assay. Expression level of Eag1 protein was detected by Western blot, and mRNA expressions of p21, p27, CyclinE1 and CDK2 were determined by reverse transcription-polymerase chain reaction.
RESULTS: After treatment with imipramine in HT-29 cells at 24, 48 and 72 h, IC50 were 43, 32 and 22 μmol/L, respectively. Cell viability decreased dose-dependently and time-dependently after treatment with imiprmince in HT 29 cells. Cell cycle arrested during the G0/G1 phase accompanied by the induction of apoptosis in a dose-dependent manner. With imipramine increasing, HT-29 cells apoptosis index gradually increased (P < 0.01). Expression level of Eag1 protein was decreased in a dose-dependent manner (P < 0.05). The p21 mRNA and p27 mRNA were up-regulated (P < 0.05), and CDK2 mRNA and CyclinE1 mRNA were suppressed in imipramine-treated HT-29 cells in a dose-dependent manner (P < 0.05).
CONCLUSION: Imipramine, a non-specific inhibitor of Eag1 potassium channel, induces cell growth inhibition, cell-cycle arrest and apoptosis in HT-29 cells through up-regulation of p27 and/or p21.
Collapse
|
90
|
|
91
|
|
92
|
Ding XW, Luo HS, Jin X, Yan JJ, Ai YW. Aberrant expression of Eag1 potassium channels in gastric cancer patients and cell lines. Med Oncol 2008; 24:345-50. [PMID: 17873312 DOI: 10.1007/s12032-007-0015-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 11/30/1999] [Accepted: 12/24/2006] [Indexed: 01/22/2023]
Abstract
Recently, an interesting relationship between potassium channels and cancer has evolved. The aim of this study is to investigate expression of Eag1 potassium channel in gastric cancer and its role in cancer cells growth. The expression of Eag1 for gasric cancer patients and cell lines as well as gastric adenoma was investigated by immunohistochemistry and reverse transcription polymerase chain reaction. In addition, imipramine was used to identify the involvement of Eag1 in the growth of SGC-7901 and BGC-823 cells. Frequency of positive expression of Eag1 protein was 70.5% (67/95) and Eag1 mRNA was 68.2% (15/22) in gastric cancer primary tissues. Eag1 mRNA was positively expressed in two gastric cell lines. Eag1 protein and mRNA were negatively expressed in paired non-cancerous matched tissues and 5 cases of adenoma tissues. The expression level of Eag1 protein was associated with lymph node metastasis (P = 0.049) and stage (P = 0.039), but had no correlation with sex, age, differentiation grades, and other organs metastases. Imipramine significantly inhibited the proliferation of SGC-7901 and BGC-823 cells at 12 h and 24 h detected by cells number counting and MTT assay (P < 0.01). The study indicates Eag1 is aberrantly expressed in gastric cancer tissues and cell lines and associated with cancer lymph node metastasis and stage and play an important role in the proliferation of gastric cancer cells.
Collapse
Affiliation(s)
- Xiang-Wu Ding
- Department of Gastroenterology, Renmin Hospital, Wuhan University, Wuhan 430060, P.R., China.
| | | | | | | | | |
Collapse
|
93
|
Ding XW, Luo HS. Ether à go-go potassium channel and malignant tumors. Shijie Huaren Xiaohua Zazhi 2007; 15:2772-2779. [DOI: 10.11569/wcjd.v15.i26.2772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recently, increasing evidence from cell biology and pharmacology demonstrates that cancer cells exhibit ion channel expression patterns, ion conductances and electric properties that are very different from those of resting cells. These peculiar properties are functionally involved in cancer pathogenesis. In particular, because of its oncogenic properties, distribution, modulation and pharmacology, human ether à go-go potassium channel (Eag1, Kv10.1, KCNH1) is considered a critical ion channel-encoding gene involved in the establishment and maintenance of neoplastic growth. This review summarizes most of the findings regarding Eag1 channels and malignant tumors, focusing on cellular mechanisms, mRNA and protein expression in tissues, oncogenic properties, modulation and pharmacology.
Collapse
|
94
|
Gómez-Varela D, Zwick-Wallasch E, Knötgen H, Sánchez A, Hettmann T, Ossipov D, Weseloh R, Contreras-Jurado C, Rothe M, Stühmer W, Pardo LA. Monoclonal antibody blockade of the human Eag1 potassium channel function exerts antitumor activity. Cancer Res 2007; 67:7343-9. [PMID: 17671204 DOI: 10.1158/0008-5472.can-07-0107] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The potassium channel ether à go-go has been directly linked to cellular proliferation and transformation, although its physiologic role(s) are as of yet unknown. The specific blockade of human Eag1 (hEag1) may not only allow the dissection of the role of the channel in distinct physiologic processes, but because of the implication of hEag1 in tumor biology, it may also offer an opportunity for the treatment of cancer. However, members of the potassium channel superfamily are structurally very similar to one another, and it has been notoriously difficult to obtain specific blockers for any given channel. Here, we describe and validate the first rational design of a monoclonal antibody that selectively inhibits a potassium current in intact cells. Specifically blocking hEag1 function using this antibody inhibits tumor cell growth both in vitro and in vivo. Our data provide a proof of concept that enables the generation of functional antagonistic monoclonal antibodies against ion channels with therapeutic potential. The particular antibody described here, as well as the technique developed to make additional functional antibodies to Eag1, makes it possible to evaluate the potential of the channel as a target for cancer therapy.
Collapse
|
95
|
Borsook D, Pendse G, Aiello-Lammens M, Glicksman M, Gostic J, Sherman S, Korn J, Shaw M, Stewart K, Gostic R, Bazes S, Hargreaves R, Becerra L. CNS response to a thermal stressor in human volunteers and rats may predict the clinical utility of analgesics. Drug Dev Res 2007. [DOI: 10.1002/ddr.20163] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
96
|
Spitzner M, Ousingsawat J, Scheidt K, Kunzelmann K, Schreiber R. Voltage-gated K+ channels support proliferation of colonic carcinoma cells. FASEB J 2006; 21:35-44. [PMID: 17135369 DOI: 10.1096/fj.06-6200com] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Plasma membrane potassium (K+) channels are required for cell proliferation. Evidence is growing that K+ channels play a central role in the development and growth of human cancer. Here we examine the contribution and the mechanism by which K+ channels control proliferation of T84 human colonic carcinoma cells. Numerous K+ channels are expressed in T84 cells, but only voltage-gated K+ (Kv) channels influenced proliferation. A number of Kv channel inhibitors reduced DNA synthesis and cell number, without exerting apoptotic or toxic effects. Expression of several Kv channels, such as EagI, Kv 3.4 and Kv 1.5, was detected in patch clamp experiments and in fluorescence-based assays using a voltage sensitive dye. The contribution of EagI channels to proliferation was confirmed by siRNA, which abolished EagI activity and inhibited cell growth. Inhibition of Kv channels did not interfere with the ability of T84 cells to regulate their cell vol, but it restricted intracellular pH regulation. In addition, inhibitors of Kv channels, as well as siRNA for EagI, attenuated intracellular Ca2+ signaling. The data suggest that Kv channels control proliferation of colonic cancer cells by affecting intracellular pH and Ca2+ signaling.
Collapse
Affiliation(s)
- Melanie Spitzner
- Institut für Physiologie, Universität Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | | | | | | | | |
Collapse
|
97
|
Su S, Ohno Y, Lossin C, Hibino H, Inanobe A, Kurachi Y. Inhibition of Astroglial Inwardly Rectifying Kir4.1 Channels by a Tricyclic Antidepressant, Nortriptyline. J Pharmacol Exp Ther 2006; 320:573-80. [PMID: 17071817 DOI: 10.1124/jpet.106.112094] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The inwardly rectifying K(+) (Kir) channel Kir4.1 is responsible for astroglial K(+) buffering. We examined the effects of nortriptyline, a tricyclic antidepressant (TCA), on Kir4.1 channel currents heterologously expressed in HEK293T cells, using a whole-cell patch-clamp technique. Nortriptyline (3-300 microM) reversibly inhibited Kir4.1 currents in a concentration-dependent manner, whereas it marginally affected neuronal Kir2.1 currents. The inhibition of Kir4.1 channels by nortriptyline depended on the voltage difference from the K(+) equilibrium potential (E(K)), with greater potency at more positive potentials. Blocking kinetics of the drug could be described by first-order kinetics, where dissociation of the drug slowed down and association accelerated as the membrane was depolarized. The dissociation constant (K(d)) of nortriptyline for Kir4.1 inhibition was 28.1 microM at E(K). Other TCAs, such as amitriptyline, desipramine, and imipramine, also inhibited Kir4.1 currents in a similar voltage-dependent fashion. This study shows for the first time that nortriptyline and related TCAs cause a concentration-, voltage-, and time-dependent inhibition of astroglial K(+)-buffering Kir4.1 channels, which might be involved in therapeutic and/or adverse actions of the drugs.
Collapse
Affiliation(s)
- Suwen Su
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
98
|
Hemmerlein B, Weseloh RM, Mello de Queiroz F, Knötgen H, Sánchez A, Rubio ME, Martin S, Schliephacke T, Jenke M, Stühmer W, Pardo LA. Overexpression of Eag1 potassium channels in clinical tumours. Mol Cancer 2006; 5:41. [PMID: 17022810 PMCID: PMC1621079 DOI: 10.1186/1476-4598-5-41] [Citation(s) in RCA: 205] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Accepted: 10/05/2006] [Indexed: 01/12/2023] Open
Abstract
Background Certain types of potassium channels (known as Eag1, KCNH1, Kv10.1) are associated with the production of tumours in patients and in animals. We have now studied the expression pattern of the Eag1 channel in a large range of normal and tumour tissues from different collections utilising molecular biological and immunohistochemical techniques. Results The use of reverse transcription real-time PCR and specifically generated monoclonal anti-Eag1 antibodies showed that expression of the channel is normally limited to specific areas of the brain and to restricted cell populations throughout the body. Tumour samples, however, showed a significant overexpression of the channel with high frequency (up to 80% depending on the tissue source) regardless of the detection method (staining with either one of the antibodies, or detection of Eag1 RNA). Conclusion Inhibition of Eag1 expression in tumour cell lines reduced cell proliferation. Eag1 may therefore represent a promising target for the tailored treatment of human tumours. Furthermore, as normal cells expressing Eag1 are either protected by the blood-brain barrier or represent the terminal stage of normal differentiation, Eag1 based therapies could produce only minor side effects.
Collapse
Affiliation(s)
- Bernhard Hemmerlein
- Department of Pathology, Georg-August University, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Rüdiger M Weseloh
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Fernanda Mello de Queiroz
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
- Divisão de Farmacologia, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rua André Cavalcanti 37/3° andar, Rio de Janeiro, Brasil
| | | | - Araceli Sánchez
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - María E Rubio
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Sabine Martin
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
- DFG Research Center for the Molecular Physiology of the Brain (CMPB), Göttingen, Germany
| | | | - Marc Jenke
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
- iOnGen AG, Stiegbreite 13, 37077 Göttingen, Germany
| | - Walter Stühmer
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Luis A Pardo
- Max-Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
- iOnGen AG, Stiegbreite 13, 37077 Göttingen, Germany
| |
Collapse
|
99
|
Mello de Queiroz F, Suarez-Kurtz G, Stühmer W, Pardo LA. Ether à go-go potassium channel expression in soft tissue sarcoma patients. Mol Cancer 2006; 5:42. [PMID: 17022811 PMCID: PMC1618397 DOI: 10.1186/1476-4598-5-42] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Accepted: 10/05/2006] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The expression of the human Eag1 potassium channel (Kv10.1) is normally restricted to the adult brain, but it has been detected in both tumour cell lines and primary tumours. Our purpose was to determine the frequency of expression of Eag1 in soft tissue sarcoma and its potential clinical implications. RESULTS We used specific monoclonal antibodies to determine the expression levels of Eag1 in soft tissue sarcomas from 210 patients by immunohistochemistry. Eag1 was expressed in 71% of all tumours, with frequencies ranging from 56% (liposarcoma) to 82% (rhabdomyosarcoma). We detected differences in expression levels depending on the histological type, but no association was seen between expression of this protein and sex, age, grade or tumour size. Four cell lines derived from relevant sarcoma histological types (fibrosarcoma and rhabdomyosarcoma) were tested for Eag1 expression by real-time RT-PCR. We found all four lines to be positive for Eag1. In these cell lines, blockage of Eag1 by RNA interference led to a decrease in proliferation. CONCLUSION Eag1 is aberrantly expressed in over 70% sarcomas. In sarcoma cell lines, inhibition of Eag1 expression and/or function leads to reduced proliferation. The high frequency of expression of Eag1 in primary tumours and the restriction of normal expression of the channel to the brain, suggests the application of this protein for diagnostic or therapeutic purposes.
Collapse
Affiliation(s)
- Fernanda Mello de Queiroz
- Divisão de Farmacologia, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rua André Cavalcanti 37/3° andar, Rio de Janeiro, Brazil
- Max-Planck-Institut für Experimentelle Medizin Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Guilherme Suarez-Kurtz
- Divisão de Farmacologia, Coordenação de Pesquisa, Instituto Nacional do Câncer, Rua André Cavalcanti 37/3° andar, Rio de Janeiro, Brazil
| | - Walter Stühmer
- Max-Planck-Institut für Experimentelle Medizin Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Luis A Pardo
- Max-Planck-Institut für Experimentelle Medizin Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| |
Collapse
|
100
|
Gómez-Varela D, Contreras-Jurado C, Furini S, García-Ferreiro R, Stühmer W, Pardo LA. Different relevance of inactivation and F468 residue in the mechanisms of hEag1 channel blockage by astemizole, imipramine and dofetilide. FEBS Lett 2006; 580:5059-66. [PMID: 16949586 DOI: 10.1016/j.febslet.2006.08.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 08/14/2006] [Accepted: 08/15/2006] [Indexed: 11/30/2022]
Abstract
The relevance of a point mutation at the C-terminal end of the S6 helix (F468) and the introduction of C-type inactivation in the blockage of hEag1 channels by astemizole, imipramine and dofetilide was tested. C-type inactivation decreased block by astemizole and dofetilide but not imipramine, suggesting different binding sites in the channel. F468C mutation increased IC(50) for astemizole and imipramine but in contrast to HERG channels, only slightly for dofetilide. Together with measurements on recovery of blocking, our observations indicate that the mechanism of hEag1 blockage by each of these drugs is different, and suggest relevant structural differences between hEag1 and HERG channels.
Collapse
Affiliation(s)
- David Gómez-Varela
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Biology of Neuronal Signals, Hermann-Rein-Str. 3, 37075 Göttingen, Germany.
| | | | | | | | | | | |
Collapse
|