1
|
Abstract
To date, research on the human ether-a-go-go related gene (hERG) has focused on this potassium channel's role in cardiac repolarization and Long QT Syndrome (LQTS). However, growing evidence implicates hERG in a diversity of physiologic and pathological processes. Here we discuss these other functions of hERG, particularly their impact on diseases beyond cardiac arrhythmia.
Collapse
|
2
|
He FZ, McLeod HL, Zhang W. Current pharmacogenomic studies on hERG potassium channels. Trends Mol Med 2013; 19:227-38. [PMID: 23369369 DOI: 10.1016/j.molmed.2012.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 12/18/2012] [Accepted: 12/27/2012] [Indexed: 11/25/2022]
Abstract
Genetic polymorphisms in human ether-a-go-go-related gene (hERG) potassium channels are associated with many complex diseases and sensitivity to channel-related drugs. Genotypes may underlie different sensitivities to the same drug, and different drugs selectively repair the functional deficits caused by individual mutations. In fact, not all drugs that block hERG function have adverse effects as previously thought. This suggests that the severe adverse reactions observed clinically may only occur in subjects with a particular genotype, but to others may be safe. Similarly, a drug that is ineffective in one population may be both safe and effective in another. Therefore, detecting polymorphisms in KCNH2 encoding hERG1 is of great significance in guiding the prevention and treatment of related diseases, re-evaluating drug safety, and individualizing treatment. This article reviews current pharmacogenomic studies on hERG potassium channels to provide a reference for developing individualized treatments and evaluating their safety.
Collapse
Affiliation(s)
- Fa-Zhong He
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan 410078, PR China
| | | | | |
Collapse
|
3
|
Molecular mechanisms of pituitary endocrine cell calcium handling. Cell Calcium 2011; 51:212-21. [PMID: 22138111 DOI: 10.1016/j.ceca.2011.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/30/2011] [Accepted: 11/07/2011] [Indexed: 11/23/2022]
Abstract
Endocrine pituitary cells express numerous voltage-gated Na(+), Ca(2+), K(+), and Cl(-) channels and several ligand-gated channels, and they fire action potentials spontaneously. Depending on the cell type, this electrical activity can generate localized or global Ca(2+) signals, the latter reaching the threshold for stimulus-secretion coupling. These cells also express numerous G-protein-coupled receptors, which can stimulate or silence electrical activity and Ca(2+) influx through voltage-gated Ca(2+) channels and hormone release. Receptors positively coupled to the adenylyl cyclase signaling pathway stimulate electrical activity with cAMP, which activates hyperpolarization-activated cyclic nucleotide-regulated channels directly, or by cAMP-dependent kinase-mediated phosphorylation of K(+), Na(+), Ca(2+), and/or non-selective cation-conducting channels. Receptors that are negatively coupled to adenylyl cyclase signaling pathways inhibit spontaneous electrical activity and accompanied Ca(2+) transients predominantly through the activation of inwardly rectifying K(+) channels and the inhibition of voltage-gated Ca(2+) channels. The Ca(2+)-mobilizing receptors activate inositol trisphosphate-gated Ca(2+) channels in the endoplasmic reticulum, leading to Ca(2+) release in an oscillatory or non-oscillatory manner, depending on the cell type. This Ca(2+) release causes a cell type-specific modulation of electrical activity and intracellular Ca(2+) handling.
Collapse
|
4
|
Glassmeier G, Hempel K, Wulfsen I, Bauer CK, Schumacher U, Schwarz JR. Inhibition of HERG1 K+ channel protein expression decreases cell proliferation of human small cell lung cancer cells. Pflugers Arch 2011; 463:365-76. [PMID: 22075718 PMCID: PMC3261411 DOI: 10.1007/s00424-011-1045-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/12/2011] [Accepted: 10/14/2011] [Indexed: 01/30/2023]
Abstract
HERG (human ether-à-go-go-related gene) K+ currents fulfill important ionic functions in cardiac and other excitable cells. In addition, HERG channels influence cell growth and migration in various types of tumor cells. The mechanisms underlying these functions are still not resolved. Here, we investigated the role of HERG channels for cell growth in a cell line (SW2) derived from small cell lung cancer (SCLC), a malignant variant of lung cancer. The two HERG1 isoforms (HERG1a, HERG1b) as well as HERG2 and HERG3 are expressed in SW2 cells. Inhibition of HERG currents by acute or sustained application of E-4031, a specific ERG channel blocker, depolarized SW2 cells by 10–15 mV. This result indicated that HERG K+ conductance contributes considerably to the maintenance of the resting potential of about −45 mV. Blockage of HERG channels by E-4031 for up to 72 h did not affect cell proliferation. In contrast, siRNA-induced inhibition of HERG1 protein expression decreased cell proliferation by about 50%. Reduction of HERG1 protein expression was confirmed by Western blots. HERG current was almost absent in SW2 cells transfected with siRNA against HERG1. Qualitatively similar results were obtained in three other SCLC cell lines (OH1, OH3, H82), suggesting that the HERG1 channel protein is involved in SCLC cell growth, whereas the ion-conducting function of HERG1 seems not to be important for cell growth.
Collapse
Affiliation(s)
- Günter Glassmeier
- Institut für Zelluläre und Integrative Physiologie, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Martinistr. 52, D-20246, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
5
|
Abstract
The human ether-a-go-go-related gene potassium channel (hERG, Kv11.1, KCNH2) has an essential role in cardiac action potential repolarization. Electrical dysfunction of the voltage-sensitive ion channel is associated with potentially lethal ventricular arrhythmias in humans. hERG K+ channels are also expressed in a variety of cancer cells where they control cell proliferation and apoptosis. In this review, we discuss molecular mechanisms of hERG-associated cell cycle regulation and cell death. In addition, the significance of hERG K+ channels as future drug target in anticancer therapy is highlighted.
Collapse
|
6
|
Wang Y, Zhang Y, Yang L, Cai B, Li J, Zhou Y, Yin L, Yang L, Yang BF, Lu YJ. Arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells through activation of caspase-3 and inhibition of HERG channels. Exp Ther Med 2011; 2:481-486. [PMID: 22977528 DOI: 10.3892/etm.2011.224] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 02/28/2011] [Indexed: 01/27/2023] Open
Abstract
Arsenic trioxide (As(2)O(3)) has been widely used to treat patients with acute promyelocytic leukemia and has also been shown to exhibit therapeutic effects on various types of solid tumors, including gastric cancer and lung carcinoma. Breast cancer is a type of solid tumor whose incidence has been increasing for many years. The present study was designed to investigate the effects of As(2)O(3) on the human breast cancer cell line MCF-7, and to explore its potential mechanisms. The MTT assay demonstrated that As(2)O(3) decreased the cellular viability of MCF-7 cells in a concentration-dependent manner. Morphological observation, the TUNEL assay and flow cytometric analysis revealed that apoptosis was involved in the process. An assay for caspase-3 activity suggested that the apoptosis was mediated through caspase-3 activation. Further investigation indicated that protein levels of the human ether-a-go-go-related gene (HERG) were markedly downregulated by As(2)O(3). Taken together, the results indicate that arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells at least in part through the activation of caspase-3 and the decrease in HERG expression.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine and Pharmaceutics, and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
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
|
8
|
Abstract
Endocrine pituitary cells are neuronlike; they express numerous voltage-gated sodium, calcium, potassium, and chloride channels and fire action potentials spontaneously, accompanied by a rise in intracellular calcium. In some cells, spontaneous electrical activity is sufficient to drive the intracellular calcium concentration above the threshold for stimulus-secretion and stimulus-transcription coupling. In others, the function of these action potentials is to maintain the cells in a responsive state with cytosolic calcium near, but below, the threshold level. Some pituitary cells also express gap junction channels, which could be used for intercellular Ca(2+) signaling in these cells. Endocrine cells also express extracellular ligand-gated ion channels, and their activation by hypothalamic and intrapituitary hormones leads to amplification of the pacemaking activity and facilitation of calcium influx and hormone release. These cells also express numerous G protein-coupled receptors, which can stimulate or silence electrical activity and action potential-dependent calcium influx and hormone release. Other members of this receptor family can activate calcium channels in the endoplasmic reticulum, leading to a cell type-specific modulation of electrical activity. This review summarizes recent findings in this field and our current understanding of the complex relationship between voltage-gated ion channels, ligand-gated ion channels, gap junction channels, and G protein-coupled receptors in pituitary cells.
Collapse
Affiliation(s)
- Stanko S Stojilkovic
- Program in Developmental Neuroscience, National Institute of Child Health and Human Development, National Institutes of Health, Building 49, Room 6A-36, 49 Convent Drive, Bethesda, Maryland 20892-4510, USA.
| | | | | |
Collapse
|
9
|
Ding XW, Yang WB, Gao S, Wang W, Li Z, Hu WM, Li JJ, Luo HS. Prognostic significance of hERG1 expression in gastric cancer. Dig Dis Sci 2010; 55:1004-10. [PMID: 19495974 DOI: 10.1007/s10620-009-0834-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 04/30/2009] [Indexed: 01/12/2023]
Abstract
Previous studies have demonstrated that human ether-à-go-go-related potassium channel (hERG1) is highly expressed in many tumor cell lines, as well as in primary human cancers, and, hence, have a critical role in cell cycle progress and proliferation. In this study, hERG1 expression was investigated in gastric cancer by immunohistochemistry and/or reverse transcription polymerase chain reaction (RT-PCR). It was discovered that hERG1, which was negatively expressed in surrounding non-tumor tissues, switched to aberrantly positive expression in gastric cancer. Statistically, there were significant differences in hERG1 protein expression according to factors such as serosal invasion, venous invasion, lymph node metastases, other organ metastases, and stage. The mean survival time for the hERG1-positive expression group was significantly shorter than the negative group, the survival rates for the positive group were significantly lower than the negative group, and hERG1 expression was found to be an independent prognostic factor. In summary, hERG1 channel was proved to be a potential biomarker for gastric cancer invasion and survival.
Collapse
Affiliation(s)
- Xiang-Wu Ding
- Department of Gastroenterology, Xiangfan Central Hospital, Xiangfan, 441021, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Sanguinetti MC. HERG1 channelopathies. Pflugers Arch 2009; 460:265-76. [PMID: 20544339 DOI: 10.1007/s00424-009-0758-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 11/03/2009] [Accepted: 11/06/2009] [Indexed: 01/08/2023]
Abstract
Human ether a go-go-related gene type 1 (hERG1) K+ channels conduct the rapid delayed rectifier K+ current and mediate action potential repolarization in the heart. Mutations in KCNH2 (the gene that encodes hERG1) causes LQT2, one of the most common forms of long QT syndrome, a disorder of cardiac repolarization that predisposes affected subjects to ventricular arrhythmia and increases the risk of sudden cardiac death. Hundreds of LQT2-associated mutations have been described, and most cause a loss of function by disrupting subunit folding, assembly, or trafficking of the channel to the cell surface. Loss-of-function mutations in hERG1 channels have also recently been implicated in epilepsy. A single gain-of-function mutation has been described that causes short QT syndrome and cardiac arrhythmia. In addition, up-regulation of hERG1 channel expression has been demonstrated in specific tumors and has been associated with skeletal muscle atrophy in mice.
Collapse
Affiliation(s)
- Michael C Sanguinetti
- Department of Physiology, Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, 95 South 2000 East, Salt Lake, UT 84112, USA.
| |
Collapse
|
11
|
Chen BS, Lo YC, Peng H, Hsu TI, Wu SN. Effects of ranolazine, a novel anti-anginal drug, on ion currents and membrane potential in pituitary tumor GH(3) cells and NG108-15 neuronal cells. J Pharmacol Sci 2009; 110:295-305. [PMID: 19609066 DOI: 10.1254/jphs.09018fp] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Ranolazine, a piperazine derivative, is currently approved for the treatment of chronic angina. However, its ionic mechanisms in other types of cells remain unclear, although it is thought to be a selective blocker of late Na(+) current. This study was conducted to evaluate the possible effects of ranolazine on Na(+) current (I(Na)), L-type Ca(2+) current (I(Ca,L)), inwardly rectifying K(+) current (I(K(IR))), delayed-rectifier K(+) current (I(K(DR))), and Ca(2+)-activated K(+) current (I(K(Ca))) in pituitary tumor (GH(3)) cells. Ranolazine depressed the transient and late components of I(Na) with different potencies. This drug exerted an inhibitory effect on I(K(IR)) with an IC(50) value of 0.92 microM, while it slightly inhibited I(K(DR)) and I(K(Ca)). It shifted the steady-state activation curve of I(K(IR)) to more positive potentials with no change in the gating charge of the channel. Ranolazine (30 microM) also reduced the activity of large-conductance Ca(2+)-activated K(+) channels in HEK293T cells expressing alpha-hSlo. Under current-clamp conditions, low concentrations (e.g., 1 microM) of ranolazine increased the firing of action potentials, while at high concentrations (>or=10 microM), it diminished the firing discharge. The exposure to ranolazine also suppressed I(Na) and I(K(IR)) effectively in NG108-15 neuronal cells. Our study provides evidence that ranolazine could block multiple ion currents such as I(Na) and I(K(IR)) and suggests that these actions may contribute to some of the functional activities of neurons and endocrine or neuroendocrine cells in vivo.
Collapse
Affiliation(s)
- Bing-Shuo Chen
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Taiwan
| | | | | | | | | |
Collapse
|
12
|
Hardy AB, Fox JEM, Giglou PR, Wijesekara N, Bhattacharjee A, Sultan S, Gyulkhandanyan AV, Gaisano HY, MacDonald PE, Wheeler MB. Characterization of Erg K+ channels in alpha- and beta-cells of mouse and human islets. J Biol Chem 2009; 284:30441-52. [PMID: 19690348 DOI: 10.1074/jbc.m109.040659] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated eag-related gene (Erg) K(+) channels regulate the electrical activity of many cell types. Data regarding Erg channel expression and function in electrically excitable glucagon and insulin producing cells of the pancreas is limited. In the present study Erg1 mRNA and protein were shown to be highly expressed in human and mouse islets and in alpha-TC6 and Min6 cells alpha- and beta-cell lines, respectively. Whole cell patch clamp recordings demonstrated the functional expression of Erg1 in alpha- and beta-cells, with rBeKm1, an Erg1 antagonist, blocking inward tail currents elicited by a double pulse protocol. Additionally, a small interference RNA approach targeting the kcnh2 gene (Erg1) induced a significant decrease of Erg1 inward tail current in Min6 cells. To investigate further the role of Erg channels in mouse and human islets, ratiometric Fura-2 AM Ca(2+)-imaging experiments were performed on isolated alpha- and beta-cells. Blocking Erg channels with rBeKm1 induced a transient cytoplasmic Ca(2+) increase in both alpha- and beta-cells. This resulted in an increased glucose-dependent insulin secretion, but conversely impaired glucagon secretion under low glucose conditions. Together, these data present Erg1 channels as new mediators of alpha- and beta-cell repolarization. However, antagonism of Erg1 has divergent effects in these cells; to augment glucose-dependent insulin secretion and inhibit low glucose stimulated glucagon secretion.
Collapse
Affiliation(s)
- Alexandre B Hardy
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Functional properties of human neuronal Kv11 channels. Pflugers Arch 2009; 458:689-700. [DOI: 10.1007/s00424-009-0651-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 02/12/2009] [Indexed: 11/25/2022]
|
14
|
The role of ether-a-go-go-related gene K(+) channels in glucocorticoid inhibition of adrenocorticotropin release by rat pituitary cells. ACTA ACUST UNITED AC 2008; 152:73-8. [PMID: 18835572 DOI: 10.1016/j.regpep.2008.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 07/17/2008] [Accepted: 09/11/2008] [Indexed: 11/22/2022]
Abstract
The present study investigated the role of K(+) channels in the inhibitory effect of glucocorticoid on adrenocorticotropin (ACTH) release induced by corticotropin-releasing hormone (CRH) using cultured rat anterior pituitary cells. Apamin and charybdotoxin (CTX) did not have a significant effect on ACTH release induced by CRH (1 nM). Tetraethylammonium (TEA), a broad spectrum K(+) channel blocker, increased the ACTH response to CRH only at the highest concentration (10 mM). The exposure to 100 nM corticosterone for 60 min inhibited the CRH-induced ACTH release. Neither TEA, apamin, nor CTX affected the inhibitory effect of corticosterone. In contrast, astemizole (Ast) and E-4031, ether-a-go-go-related gene (erg) K(+) channel blockers, abolished the inhibitory effect of corticosterone on CRH-induced ACTH release (1.25+/-0.10 vs. 1.45+/-0.11 ng/well at 10 microM Ast, p>0.05, 1.71+/-0.16 vs. 1.91+/-0.32 ng/well at 10 microM E-4031, p>0.05, vehicle vs. corticosterone). RT-PCR demonstrated all three subtypes of rat-erg mRNAs in the pituitary and corticosterone increased only erg1 mRNA up to 2.47+/-0.54 fold. In conclusion, erg K(+) channels were up-regulated by glucocorticoid, and have indispensable roles in delayed glucocorticoid inhibition of CRH-induced ACTH release by rat pituitary cells.
Collapse
|
15
|
Cavarra MS, del Mónaco SM, Assef YA, Ibarra C, Kotsias BA. HERG1 currents in native K562 leukemic cells. J Membr Biol 2007; 219:49-61. [PMID: 17763876 DOI: 10.1007/s00232-007-9060-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Accepted: 06/18/2007] [Indexed: 10/22/2022]
Abstract
The human ether-a-go-go related gene (HERG1) K+ channel is expressed in neoplastic cells, in which it was proposed to play a role in proliferation, differentiation and/or apoptosis. K562 cells (a chronic myeloid leukemic human cell line) express both the full-length (herg1a) and the N-terminally truncated (herg1b) isoforms of the gene, and this was confirmed with Western blots and coimmunoprecipitation experiments. Whole-cell currents were studied with a tail protocol. Seventy-eight percent of cells showed a HERG1-like current: repolarization to voltages negative to -40 mV produced a transient peak inward tail current, characteristic of HERG1 channels. Cells were exposed to a HERG-specific channel blocker, E4031. Half-maximal inhibitory concentration (IC50) of the blocker was 4.69 nM: The kinetics of the HERG1 current in K562 cells resembled the rapid component of the native cardiac delayed rectifier current, known to be conducted by heterotetrameric HERG1 channels. Fast and slow deactivation time constants at -120 mV were 27.5 and 239.5 ms, respectively. Our results in K562 cells suggest the assembling of heterotetrameric channels, with some parameters being dominated by one of the isoforms and other parameters being intermediate. Hydrogen peroxide was shown to increase HERG1a K+ current in heterologous expression systems, which constitutes an apoptotic signal. However, we found that K562 HERG1 whole-cell currents were not activated by H2O2.
Collapse
Affiliation(s)
- María S Cavarra
- Laboratorio de Neurofisiología, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires-CONICET, C. de Malvinas 3150, Buenos Aires, 1427, Argentina
| | | | | | | | | |
Collapse
|
16
|
Rodriguez S, Gaunt TR, Day INM. Molecular genetics of human growth hormone, insulin-like growth factors and their pathways in common disease. Hum Genet 2007; 122:1-21. [PMID: 17534663 DOI: 10.1007/s00439-007-0378-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2006] [Accepted: 05/08/2007] [Indexed: 12/29/2022]
Abstract
The human growth hormone gene (GH1) and the insulin-like growth factor 1 and 2 genes (IGF1 and IGF2) encode the central elements of a key pathway influencing growth in humans. This "growth pathway" also includes transcription factors, agonists, antagonists, receptors, binding proteins, and endocrine factors that constitute an intrincate network of feedback loops. GH1 is evolutionarily coupled with other genes in linkage disequilibrium in 17q24.2, and the same applies to IGF2 in 11p15.5. In contrast, IGF1 in 12q22-24.1 is not in strong linkage disequilibrium with neighbouring genes. Knowledge of the functional architecture of these regions is important for the understanding of the combined evolution and function of GH1, IGF2 and IGF1 in relation to complex diseases. A number of mutations accounting for rare Mendelian disorders have been described in GH-IGF elements. The constellation of genes in this key pathway contains potential candidates in a number of complex diseases, including growth disorders, metabolic syndrome, diabetes (notably IGF2BP2) cardiovascular disease, and central nervous system diseases, and in longevity, aging and cancer. We review these genes and their associations with disease phenotypes, with special attention to metabolic risk traits.
Collapse
Affiliation(s)
- Santiago Rodriguez
- Bristol Genetic Epidemiology Laboratories and MRC Centre for Causal Analyses in Translational Epidemiology (CAiTE), Department of Social Medicine, University of Bristol, Canynge Hall, Whiteladies Road, Bristol, BS8 2PR, UK.
| | | | | |
Collapse
|
17
|
Zhang M, Liu XS, Diochot S, Lazdunski M, Tseng GN. APETx1 from Sea Anemone Anthopleura elegantissima Is a Gating Modifier Peptide Toxin of the Human Ether-a-go-go- Related Potassium Channel. Mol Pharmacol 2007; 72:259-68. [PMID: 17473056 DOI: 10.1124/mol.107.035840] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We studied the mechanism of action and the binding site of APETx1, a peptide toxin purified from sea anemone, on the human ether-a-go-go-related gene (hERG) channel. Similar to the effects of gating modifier toxins (hanatoxin and SGTx) on the voltage-gated potassium (Kv) 2.1 channel, APETx1 shifts the voltage-dependence of hERG activation in the positive direction and suppresses its current amplitudes elicited by strong depolarizing pulses that maximally activate the channels. The APETx1 binding site is distinctly different from that of a pore-blocking peptide toxin, BeKm-1. Mutations in the S3b region of hERG have dramatic impact on the responsiveness to APETx1: G514C potentiates whereas E518C abolishes the APETx1 effect. Restoring the negative charge at position 518 (methanethiosulfonate ethylsulfonate modification of 518C) partially restores APETx1 responsiveness, supporting an electrostatic interaction between E518 and APETx1. Among the three hERG isoforms, hERG1 and hERG3 are equally responsive to APETx1, whereas hERG2 is insensitive. The key feature seems to be an arginine residue uniquely present at the 514-equivalent position in hERG2, where the other two isoforms possess a glycine. Our data show that APETx1 is a gating modifier toxin of the hERG channel, and its binding site shares characteristics with those of gating modifier toxin binding sites on other Kv channels.
Collapse
Affiliation(s)
- M Zhang
- Department of Physiology, Medical College of Virginia, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298, USA
| | | | | | | | | |
Collapse
|
18
|
Cockerill SL, Tobin AB, Torrecilla I, Willars GB, Standen NB, Mitcheson JS. Modulation of hERG potassium currents in HEK-293 cells by protein kinase C. Evidence for direct phosphorylation of pore forming subunits. J Physiol 2007; 581:479-93. [PMID: 17363390 PMCID: PMC2075202 DOI: 10.1113/jphysiol.2006.123414] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The human ether-a-go-go related gene (hERG) potassium channel is expressed in a variety of tissues including the heart, neurons and some cancer cells. hERG channels are modulated by several intracellular signalling pathways and these provide important mechanisms for regulating cellular excitability. In this study, we investigated muscarinic modulation of hERG currents and direct phosphorylation of channel subunits expressed in HEK-293 cells at physiologically relevant temperatures by protein kinase C (PKC). Activation of G(alpha q/11)-coupled M(3)-muscarinic receptors with methacholine, reduced current amplitudes at all potentials with minor effects on the voltage dependence of activation and inactivation. The response to methacholine was insensitive to intracellular BAPTA, but was attenuated by either acute inhibition of PKC with 300 nm bisindolylmaleimide-1 (bis-1) or chronic down-regulation of PKC isoforms by 24 h pretreatment of cells with phorbol 12-myristate 13-acetate (PMA). Stimulation of PKC with 1-oleoyl 2-acetylglycerol (OAG), an analogue of diacylglycerol (DAG), mimicked the actions of muscarinic receptor stimulation. Direct phosphorylation of hERG was measured by [(32)P]orthophosphate labelling of immunoprecipitated protein with an anti-hERG antibody. Basal phosphorylation was high in unstimulated cells and further increased by OAG. The OAG dependent increase was abolished by bis-1 and down-regulation of PKC, but basal levels of phosphorylation were unchanged. Deletion of the amino-terminus of hERG prevented both the modulation of channel activity and the increase of phosphorylation by OAG. Our results are consistent with calcium and/or DAG sensitive isotypes of PKC modulating hERG currents through a mechanism that involves direct phosphorylation of sites on the amino terminus of hERG.
Collapse
Affiliation(s)
- S L Cockerill
- University of Leicester, Department of Cell Physiology and Pharmacology, Maurice Shock Medical Sciences Building, University Road, Leicester LE1 9HN, UK
| | | | | | | | | | | |
Collapse
|
19
|
Lin H, Xiao J, Luo X, Wang H, Gao H, Yang B, Wang Z. Retracted: Overexpression HERG K+ channel gene mediates cell-growth signals on activation of oncoproteins SP1 and NF-κB and inactivation of tumor suppressor Nkx3.1. J Cell Physiol 2007; 212:137-47. [PMID: 17311278 DOI: 10.1002/jcp.21015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The long QT syndrome gene human ether-a-go-go related gene (HERG) encodes a K(+) channel critical to cardiac repolarization. It peculiarly overexpresses in cancer cells of different histogenesis and promotes tumorigenesis. To decipher the molecular mechanisms for HERG overexpression, we identified and characterized the promoter region of the HERG gene, which contains cis-elements for multiple oncoproteins and tumor suppressors. Oncoprotein Sp1 was found to be essential to driving the HERG promoter thereby transcription. Another oncoprotein NF-kappaB transactivated, while tumor suppressor Nkx3.1 repressed HERG promoter activity and endogenous HERG transcription. Loss-of-function mutations in the corresponding cis-elements rendered a loss of the ability of the oncoproteins Sp1 and NF-kappaB to transactivate, and of the tumor repressor Nkx3.1 to repress, HERG transcription. Either activation of Sp1 and NF-kappaB or silencing of Nkx3.1 promoted tumor cell growth, and the effects were abrogated by HERG inhibition or knockdown, but facilitated by overexpression of HERG, indicating that HERG mediates the cell growth signals generated by activation of oncoproteins or inactivation of tumor suppressors. Binding of Sp1, NF-kappaB, and Nkx3.1 to their respective cis-elements in the HERG promoter in vitro and their presence on the HERG promoter in vivo were confirmed. Therefore, the HERG promoter region is characterized by multiple Sp1 binding sites that are responsible for transcription initiation of the HERG gene and by binding sites for multiple other oncogenes and tumor suppressor genes being important for regulating HERG expression. The HERG K(+) channel is likely a mediator of growth-promoting processes induced by oncoproteins and/or by silencing of tumor suppressors.
Collapse
Affiliation(s)
- Huixian Lin
- Research Center, Montreal Heart Institute, Montreal, Canada
| | | | | | | | | | | | | |
Collapse
|
20
|
Tseng GN, Sonawane KD, Korolkova YV, Zhang M, Liu J, Grishin EV, Guy HR. Probing the outer mouth structure of the HERG channel with peptide toxin footprinting and molecular modeling. Biophys J 2007; 92:3524-40. [PMID: 17293393 PMCID: PMC1853143 DOI: 10.1529/biophysj.106.097360] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Previous studies have shown that the unusually long S5-P linker lining human ether a-go-go related gene's (hERG's) outer vestibule is critical for its channel function: point mutations at high-impact positions here can interfere with the inactivation process and, in many cases, also reduce the pore's K+ selectivity. Because no data are available on the equivalent region in the available K channel crystal structures to allow for homology modeling, we used alternative approaches to model its three-dimensional structure. The first part of this article describes mutant cycle analysis used to identify residues on hERG's outer vestibule that interact with specific residues on the interaction surface of BeKm-1, a peptide toxin with known NMR structure and a high binding affinity to hERG. The second part describes molecular modeling of hERG's pore domain. The transmembrane region was modeled after the crystal structure of KvAP pore domain. The S5-P linker was docked to the transmembrane region based on data from previous NMR and mutagenesis experiments, as well as a set of modeling criteria. The models were further restrained by contact points between hERG's outer vestibule and the bound BeKm-1 toxin molecule deduced from the mutant cycle analysis. Based on these analyses, we propose a working model for the open conformation of the outer vestibule of the hERG channel, in which the S5-P linkers interact with the pore loops to influence ion flux through the pore.
Collapse
Affiliation(s)
- Gea-Ny Tseng
- Department of Physiology, Virginia Commonwealth University, Richmond, Virginia 23298, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Zhang M, Liu J, Jiang M, Wu DM, Sonawane K, Guy HR, Tseng GN. Interactions between charged residues in the transmembrane segments of the voltage-sensing domain in the hERG channel. J Membr Biol 2006; 207:169-81. [PMID: 16550488 DOI: 10.1007/s00232-005-0812-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2005] [Revised: 10/09/2005] [Indexed: 11/30/2022]
Abstract
Studies on voltage-gated K channels such as Shaker have shown that positive charges in the voltage-sensor (S4) can form salt bridges with negative charges in the surrounding transmembrane segments in a state-dependent manner, and different charge pairings can stabilize the channels in closed or open states. The goal of this study is to identify such charge interactions in the hERG channel. This knowledge can provide constraints on the spatial relationship among transmembrane segments in the channel's voltage-sensing domain, which are necessary for modeling its structure. We first study the effects of reversing S4's positive charges on channel activation. Reversing positive charges at the outer (K525D) and inner (K538D) ends of S4 markedly accelerates hERG activation, whereas reversing the 4 positive charges in between either has no effect or slows activation. We then use the 'mutant cycle analysis' to test whether D456 (outer end of S2) and D411 (inner end of S1) can pair with K525 and K538, respectively. Other positive charges predicted to be able, or unable, to interact with D456 or D411 are also included in the analysis. The results are consistent with predictions based on the distribution of these charged residues, and confirm that there is functional coupling between D456 and K525 and between D411 and K538.
Collapse
Affiliation(s)
- M Zhang
- Department of Physiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Camacho J. Ether à go-go potassium channels and cancer. Cancer Lett 2006; 233:1-9. [PMID: 16473665 DOI: 10.1016/j.canlet.2005.02.016] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 02/14/2005] [Indexed: 12/28/2022]
Abstract
Ion channels play important roles in health and disease. In the last few years, an interesting relationship between potassium channels and cancer has evolved. Especially, members of the ether à go-go (EAG) potassium channels family have gained interest as research tools for detection and therapy of different cancers. This review will summarize most of the findings relating EAG channels and cancer, focusing on mRNA expression in tissues, oncogenic properties, modulation and pharmacology. Plausible scenarios on the cellular mechanisms of EAG oncogenicity will be discussed.
Collapse
Affiliation(s)
- Javier Camacho
- Pharmacology Section, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Avenida Instituto Politécnico Nacional 2508, México, DF 07360, México.
| |
Collapse
|
23
|
McKay CM, Ye J, Huizinga JD. Characterization of depolarization-evoked ERG K currents in interstitial cells of Cajal. Neurogastroenterol Motil 2006; 18:324-33. [PMID: 16553588 DOI: 10.1111/j.1365-2982.2006.00764.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Interstitial cells of Cajal (ICC) harbour the ether-a-go-go related gene (ERG) channel as shown by its characteristic rapidly deactivating current upon hyperpolarization. This property, however, does not explain the marked increase in cell excitability by ERG channel blockers, namely an increase in slow wave plateau duration and action potential generation. The objective of the present study was to characterize the depolarization-activated, E4031-sensitive ERG currents in murine ICC within a range of physiologically relevant membrane potentials. Whole cell currents were recorded from ICC isolated from murine neonatal jejunum, superfused with a physiological salt solution and with high intracellular Cs(+) to block most other K(+) currents. Upon depolarizing the cell from the resting membrane potential (approximately -60 mV) towards the region of the slow wave plateau (approximately -30 mV), significant sustained (window) current was generated between the potentials of -40 to 0 mV (maximal at -30 mV) and inhibited by the ERG specific blocker E4031. Channel activation followed by rapid inactivation produced a steady state conductance at -30 mV which was 51.6 +/- 11% of the hyperpolarization-evoked peak conductance value at -100 mV. When the cell repolarized from -30 mV, again, significant currents were generated, indicating recovery from inactivation, a typical characteristic of ERG channels. These data provide evidence that the ERG channel is of significance in the regulation of ICC excitability and provide the mechanism by which ERG channel blockade increases the slow wave duration.
Collapse
Affiliation(s)
- C M McKay
- Intestinal Disease Research Programme, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
24
|
Sarzani R, Pietrucci F, Corinaldesi C, Francioni M, Letizia C, D'Erasmo E, Dessì-Fulgheri P, Rappelli A. The functional HERG variant 897T is associated with Conn's adenoma. J Hypertens 2006; 24:479-87. [PMID: 16467651 DOI: 10.1097/01.hjh.0000209984.28735.fd] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Aldosterone secreting adenomas (aldosteronomas) have an unknown molecular origin. Ion channel currents are involved in signal transduction leading to aldosterone synthesis and secretion. HERG (human-ether-a-go-go-related gene) encodes for a potassium channel responsible for the outward rectifying delayed current and it is mutation prone. When mutated it causes most of the familial forms of both long QT and short QT syndromes. Abnormal repolarization in glomerulosa cells might increase aldosterone secretion or induce a proliferative advantage. The aims of this study were to: (1) evaluate HERG expression in aldosteronomas; (2) search for HERG somatic mutations; and (3) determine whether there is any relationship between the common HERG functional variant (A2690C, leading from lysine 897 to threonine, K897T) and aldosteronoma. DESIGN AND METHODS Aldosteronoma and blood samples from 17 patients were studied to evaluate HERG expression, full-length HERG complementary DNA sequencing, and genotyping for K897T alleles. The prevalence of HERG 897 alleles was also tested in a control population and a population consisting entirely of hypertensive individuals. RESULTS HERG was expressed in all aldosteronomas analysed. HERG somatic mutations were not detected. The 897T variant of HERG was significantly more common among patients with aldosteronoma (897T allele 41%) than in patients with moderate-severe essential hypertension (897T allele 20%, P = 0.007) or in the control population (897T allele 12%, P < 0.0001). The 897T/T genotype was present in 24% of the aldosteronoma patients versus 7% (P = 0.040) and 3% (P = 0.001) in essential hypertension and in the control population, respectively. When the chi test was performed considering the three groups together, the significance was similar (for alleles P < 0.0001 and for genotypes P = 0.004). CONCLUSION The common functional HERG variant 897T may predispose to the development of aldosteronoma.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Department of Internal Medicine, Università Politecnica delle Marche, Ancona, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Gordon RD. Can mutations in ion channel genes cause endocrine dysfunction? J Hypertens 2006; 24:441-2. [PMID: 16467644 DOI: 10.1097/01.hjh.0000209977.90617.f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Masi A, Becchetti A, Restano-Cassulini R, Polvani S, Hofmann G, Buccoliero AM, Paglierani M, Pollo B, Taddei GL, Gallina P, Di Lorenzo N, Franceschetti S, Wanke E, Arcangeli A. hERG1 channels are overexpressed in glioblastoma multiforme and modulate VEGF secretion in glioblastoma cell lines. Br J Cancer 2005; 93:781-92. [PMID: 16175187 PMCID: PMC2361632 DOI: 10.1038/sj.bjc.6602775] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Recent studies have led to considerable advancement in our understanding of the molecular mechanisms that underlie the relentless cell growth and invasiveness of human gliomas. Partial understanding of these mechanisms has (1) improved the classification for gliomas, by identifying prognostic subgroups, and (2) pointed to novel potential therapeutic targets. Some classes of ion channels have turned out to be involved in the pathogenesis and malignancy of gliomas. We studied the expression and properties of K+ channels in primary cultures obtained from surgical specimens: human ether a gò-gò related (hERG)1 voltage-dependent K+ channels, which have been found to be overexpressed in various human cancers, and human ether a gò-gò-like 2 channels, that share many of hERG1's biophysical features. The expression pattern of these two channels was compared to that of the classical inward rectifying K+ channels, IRK, that are widely expressed in astrocytic cells and classically considered a marker of astrocytic differentiation. In our study, hERG1 was found to be specifically overexpressed in high-grade astrocytomas, that is, glioblastoma multiforme (GBM). In addition, we present evidence that, in GBM cell lines, hERG1 channel activity actively contributes to malignancy by promoting vascular endothelial growth factor secretion, thus stimulating the neoangiogenesis typical of high-grade gliomas. Our data provide important confirmation for studies proposing the hERG1 channel as a molecular marker of tumour progression and a possible target for novel anticancer therapies.
Collapse
Affiliation(s)
- A Masi
- Department of Experimental Pathology and Oncology, University of Firenze, Viale GB Morgagni, 50, 50134 Firenze, Italy
| | - A Becchetti
- Department of Biotechnology and Biosciences, University of Milano Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - R Restano-Cassulini
- Department of Biotechnology and Biosciences, University of Milano Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - S Polvani
- Department of Experimental Pathology and Oncology, University of Firenze, Viale GB Morgagni, 50, 50134 Firenze, Italy
| | - G Hofmann
- Department of Experimental Pathology and Oncology, University of Firenze, Viale GB Morgagni, 50, 50134 Firenze, Italy
| | - A M Buccoliero
- Department of Human Pathology and Oncology, University of Firenze, Viale GB Morgagni, 88, 50134 Firenze, Italy
| | - M Paglierani
- Department of Human Pathology and Oncology, University of Firenze, Viale GB Morgagni, 88, 50134 Firenze, Italy
| | - B Pollo
- Istituto Neurologico Besta, Via Celoria, 11, 20133 Milano, Italy
| | - G L Taddei
- Department of Human Pathology and Oncology, University of Firenze, Viale GB Morgagni, 88, 50134 Firenze, Italy
| | - P Gallina
- Department of Neurosurgery, University of Firenze, Largo P Palagi 1, 50139, Firenze, Italy
| | - N Di Lorenzo
- Department of Neurosurgery, University of Firenze, Largo P Palagi 1, 50139, Firenze, Italy
| | - S Franceschetti
- Istituto Neurologico Besta, Via Celoria, 11, 20133 Milano, Italy
| | - E Wanke
- Department of Biotechnology and Biosciences, University of Milano Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - A Arcangeli
- Department of Experimental Pathology and Oncology, University of Firenze, Viale GB Morgagni, 50, 50134 Firenze, Italy
- Department of Experimental Pathology and Oncology, University of Firenze, Viale GB Morgagni, 50, 50134 Firenze, Italy. E-mail:
| |
Collapse
|
27
|
Cockerill SL, Mitcheson JS. Direct block of human ether-a-go-go-related gene potassium channels by caffeine. J Pharmacol Exp Ther 2005; 316:860-8. [PMID: 16227470 DOI: 10.1124/jpet.105.094755] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The human ether-a-go-go-related gene (hERG) potassium channel is expressed in a variety of cell types, including neurons, tumor cells, and cardiac myocytes. In the heart, it is important for repolarization of the cardiac action potential. Attenuation of hERG current can cause long QT syndrome and cardiac arrhythmias such as torsades de pointes. Caffeine is frequently used as a pharmacological tool to study calcium-dependent transduction pathways in cellular preparations. It raises cytosolic calcium by opening ryanodine receptors and may also inhibit phosphodiesterases to increase cytosolic cAMP. In this study, we show 5 mM caffeine rapidly and reversibly attenuates hERG currents expressed in human embryonic kidney 293 cells to 61.1 +/- 2.2% of control. Caffeine-dependent inhibition of hERG current is not altered by raising cAMP with forskolin, buffering cytosolic calcium with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, or inhibition of protein kinase C. Thus, the effects of caffeine are unlikely to be mediated by cAMP or intracellular calcium-dependent mechanisms. Further experiments showed caffeine directly blocks hERG in an open state-dependent manner. Furthermore, caffeine inhibition is greatly reduced by the pore mutants Y562A and F656A hERG, which disrupt block of most previously tested hERG antagonists. Thus, caffeine attenuates hERG currents by binding to a drug receptor located within the inner cavity of the channel. Dietary intake of caffeine is unlikely to cause long QT syndrome because plasma concentrations do not reach sufficiently high levels to significantly inhibit hERG currents. However, the effects of caffeine have implications for its use in examining calcium-dependent pathways in cellular preparations expressing hERG.
Collapse
Affiliation(s)
- S L Cockerill
- Department of Cell Physiology and Pharmacology, University of Leicester, UK
| | | |
Collapse
|
28
|
Patt S, Preussat K, Beetz C, Kraft R, Schrey M, Kalff R, Schönherr K, Heinemann SH. Expression of ether à go-go potassium channels in human gliomas. Neurosci Lett 2005; 368:249-53. [PMID: 15364405 DOI: 10.1016/j.neulet.2004.07.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2003] [Revised: 05/06/2004] [Accepted: 05/06/2004] [Indexed: 11/29/2022]
Abstract
Ether à go-go (EAG) K(+) channels have been shown to be involved in tumor generation and malignant growth. Gliomas have not been investigated thus far. Using RT-PCR we investigated healthy human brain and human gliomas of different subtypes and malignancy grades for the expression of human EAG1 and eag-related gene (ERG) 1 channels. mRNA of both channels was detected in all tissues. Expression was strong in normal brain, moderate in high-grade and high in low-grade gliomas. Our findings suggest a differential expression of hEAG1 and hERG1 in gliomas depending on the malignancy grade and nature of the tumor cells. However, the hypothesis that EAG channels are related to the oncogenic process itself is only partly supported by this study.
Collapse
Affiliation(s)
- Stephan Patt
- Institute of Pathology (Neuropathology), Friedrich Schiller University Jena, Bachstr. 18, D-07740 Jena, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Kim I, Boyle KM, Carroll JL. Postnatal development of E-4031-sensitive potassium current in rat carotid chemoreceptor cells. J Appl Physiol (1985) 2004; 98:1469-77. [PMID: 15591286 DOI: 10.1152/japplphysiol.01254.2003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The O2 sensitivity of dissociated type I cells from rat carotid body increases with age until approximately 14-16 days. Hypoxia-induced depolarization appears to be mediated by an O2-sensitive K+ current, but other K+ currents may modulate depolarization. We hypothesized that membrane potential may be stabilized in newborn type I cells by human ether-a-go-go-related gene (HERG)-like K+ currents that inhibit hypoxia-induced depolarization and that a decrease in this current with age could underlie, in part, the developmental increase in type I cell depolarization response to hypoxia. In dissociated type I cells from 0- to 1- and 11- to 16-day-old rats, using perforated patch-clamp and 70 mM K+ extracellular solution, we measured repolarization-induced inward K+ tail currents in the absence and presence of E-4031, a specific HERG channel blocker. This allowed isolation of the E-4031-sensitive HERG-like current. E-4031-sensitive peak currents in type I cells from 0- to- 1-day-old rats were 2.5-fold larger than in cells from 11- to 16-day-old rats. E-4031-sensitive current density in newborn type I cells was twofold greater than in cells from 11- to 16-day-old rats. Under current clamp conditions, E-4031 enhanced hypoxia-induced depolarization in type I cells from 0- to- 1-day-old but not 11- to 16-day-old rats. With use of fura 2 to measure intracellular Ca2+, E-4031 increased the cytosolic Ca2+ concentration response to anoxia in cells from 0- to- 1-day-old but not cells from 11- to 16-day-old rats. E-4031-sensitive K+ currents are present in newborn carotid body type I cells and decline with age. These findings are consistent with a role for E-4031-sensitive K+ current, and possibly HERG-like K+ currents, in the type I cell hypoxia response maturation.
Collapse
Affiliation(s)
- Insook Kim
- Pediatric Pulmonary Division, Arkansas Children's Hospital, Slot 512-17, 800 Marshall St., Little Rock, AR 72202, USA
| | | | | |
Collapse
|
30
|
Abstract
Ether-à-go-go-related gene (erg) channels are voltage-dependent K+ channels mediating inward-rectifying K+ currents because of their peculiar gating kinetics. These characteristics are essential for repolarization of the cardiac action potential. Inherited and acquired malfunctioning of erg channels may lead to the long QT-syndrome. However, erg currents have also been recorded in many other excitable cells, like smooth muscle fibres of the gastrointestinal tract, neuroblastoma cells or neuroendocrine cells. In these cells erg currents contribute to the maintenance of the resting potential. Changes in the resting potential are related to cell-specific functions like increase in hormone secretion, frequency adaptation or increase in contractility.
Collapse
Affiliation(s)
- Jürgen R Schwarz
- Institut für Angewandte Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| | | |
Collapse
|