1
|
Forzisi E, Sesti F. Non-conducting functions of ion channels: The case of integrin-ion channel complexes. Channels (Austin) 2022; 16:185-197. [PMID: 35942524 PMCID: PMC9364710 DOI: 10.1080/19336950.2022.2108565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Started as an academic curiosity more than two decades ago, the idea that ion channels can regulate cellular processes in ways that do not depend on their conducting properties (non-ionic functions) gained traction and is now a flourishing area of research. Channels can regulate physiological processes including actin cytoskeletal remodeling, cell motility, excitation-contraction coupling, non-associative learning and embryogenesis, just to mention some, through non-ionic functions. When defective, non-ionic functions can give rise to channelopathies involved in cancer, neurodegenerative disease and brain trauma. Ion channels exert their non-ionic functions through a variety of mechanisms that range from physical coupling with other proteins, to possessing enzymatic activity, to assembling with signaling molecules. In this article, we take stock of the field and review recent findings. The concept that emerges, is that one of the most common ways through which channels acquire non-ionic attributes, is by assembling with integrins. These integrin-channel complexes exhibit broad genotypic and phenotypic heterogeneity and reveal a pleiotropic nature, as they appear to be capable of influencing both physiological and pathological processes.
Collapse
Affiliation(s)
- Elena Forzisi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| |
Collapse
|
2
|
Chioccioli Altadonna G, Montalbano A, Iorio J, Becchetti A, Arcangeli A, Duranti C. The Interaction between hERG1 and β1 Integrins Modulates hERG1 Current in Different Pathological Cell Models. MEMBRANES 2022; 12:1162. [PMID: 36422154 PMCID: PMC9698864 DOI: 10.3390/membranes12111162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 06/16/2023]
Abstract
Ion channels are implicated in various diseases, including cancer, in which they modulate different aspects of cancer progression. In particular, potassium channels are often aberrantly expressed in cancers, a major example being provided by hERG1. The latter is generally complexed with β1 integrin in tumour cells, and such a molecular complex represents a new druggable hub. The present study focuses on the characterization of the functional consequences of the interaction between hERG1 and β1 integrins on different substrates over time. To this purpose, we studied the interplay alteration on the plasma membrane through patch clamp techniques in a cellular model consisting of human embryonic kidney (HEK) cells stably transfected with hERG1 and in a cancer cell model consisting of SH-SY5Y neuroblastoma cells, endogenously expressing the channel. Cells were seeded on different substrates known to stimulate β1 integrins, such as fibronectin (FN) for HEK-hERG1 and laminin (LMN) for SH-SY5Y. In HEK cells stably overexpressing hERG1, we observed a hERG1 current density increase accompanied by Vrest hyperpolarization after cell seeding onto FN. Notably, a similar behaviour was shown by SH-SY5Y neuroblastoma cells plated onto LMN. Interestingly, we did not observe this phenomenon when plating the cells on substrates such as Bovine Serum Albumin (BSA) or Polylysine (PL), thus suggesting a crucial involvement of ECM proteins as well as of β1 integrin activation.
Collapse
Affiliation(s)
| | - Alberto Montalbano
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Jessica Iorio
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Claudia Duranti
- Department of Experimental and Clinical Medicine, University of Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| |
Collapse
|
3
|
Axonal TAU Sorting Requires the C-terminus of TAU but is Independent of ANKG and TRIM46 Enrichment at the AIS. Neuroscience 2021; 461:155-171. [PMID: 33556457 DOI: 10.1016/j.neuroscience.2021.01.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/26/2021] [Accepted: 01/31/2021] [Indexed: 01/06/2023]
Abstract
Somatodendritic missorting of the axonal protein TAU is a hallmark of Alzheimer's disease and related tauopathies. Rodent primary neurons and iPSC-derived neurons are used for studying mechanisms of neuronal polarity, including TAU trafficking. However, these models are expensive, time-consuming, and/or require the killing of animals. In this study, we tested four differentiation procedures to generate mature neuron cultures from human SH-SY5Y neuroblastoma cells and assessed the TAU sorting capacity. We show that SH-SY5Y-derived neurons, differentiated with sequential RA/BDNF treatment, are suitable for investigating axonal TAU sorting. These human neurons show pronounced neuronal polarity, axodendritic outgrowth, expression of the neuronal maturation markers TAU and MAP2, and, importantly, efficient axonal sorting of endogenous and transfected human wild-type TAU, similar to mouse primary neurons. We demonstrate that the N-terminal half of TAU is not sufficient for axonal targeting, as a C-terminus-lacking construct (N-term-TAUHA) is not axonally enriched in both neuronal cell models. Importantly, SH-SY5Y-derived neurons do not show the formation of a classical axon initial segment (AIS), indicated by the lack of ankyrin G (ANKG) and tripartite motif-containing protein 46 (TRIM46) at the proximal axon, which suggests that successful axonal TAU sorting is independent of classical AIS formation. Taken together, our results provide evidence that (i) SH-SY5Y-derived neurons are a valuable human neuronal cell model for studying TAU sorting readily accessible at low cost and without animal need, and that (ii) efficient axonal TAU targeting is independent of ANKG or TRIM46 enrichment at the proximal axon in these neurons.
Collapse
|
4
|
Şahin M, Öncü G, Yılmaz MA, Özkan D, Saybaşılı H. Transformation of SH-SY5Y cell line into neuron-like cells: Investigation of electrophysiological and biomechanical changes. Neurosci Lett 2021; 745:135628. [PMID: 33440235 DOI: 10.1016/j.neulet.2021.135628] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
SH-SY5Y human neuroblastoma cells are commonly used as neuronal models. Here, we examined different aspects of SH-SY5Y cell differentiation. Various differentiation protocols have been proposed previously, including treatments with retinoic acid, brain-derived neurotrophic factor (BDNF), cholesterol and oestradiol. We examined undifferentiated SH-SY5Y cells (UNDIFF); cells differentiated by the treatment with retinoic acid (RA); retinoic acid + BDNF (RB); and retinoic acid + BDNF + cholesterol + oestradiol (RBCE). We performed whole-cell patch-clamp recordings from these cells and nanomechanically characterised them by using atomic force microscopy (AFM). Our results indicated that Na+ currents become most pronounced in the differentiated RB cells, whereas UNDIFF SH-SY5Y cells had significantly larger K+ currents, which is a characteristic feature of cancer cells. AFM observations of these two groups showed that Young's moduli of SH-SY5Y cells increased threefold with differentiation. Furthermore, we showed a direct relationship between Na+ channel activity and elasticity in these cells. We conclude that SH-SY5Y human neuroblastoma cells should be used as a neuronal model only when they are differentiated by the treatment with retinoic acid and BDNF.
Collapse
Affiliation(s)
- Meryem Şahin
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Gül Öncü
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Mustafa Alper Yılmaz
- Department of Mechanical Engineering, National Defense University, Naval Academy, Istanbul, Turkey
| | - Doğuş Özkan
- Department of Mechanical Engineering, National Defense University, Naval Academy, Istanbul, Turkey
| | - Hale Saybaşılı
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey.
| |
Collapse
|
5
|
Cholinergic Differentiation of Human Neuroblastoma SH-SY5Y Cell Line and Its Potential Use as an In vitro Model for Alzheimer's Disease Studies. Mol Neurobiol 2019; 56:7355-7367. [PMID: 31037648 DOI: 10.1007/s12035-019-1605-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
Abstract
Cholinergic transmission is critical to high-order brain functions such as memory, learning, and attention. Alzheimer's disease (AD) is characterized by cognitive decline associated with a specific degeneration of cholinergic neurons. No effective treatment to prevent or reverse the symptoms is known. Part of this might be due to the lack of in vitro models that effectively mimic the relevant features of AD. Here, we describe the characterization of an AD in vitro model using the SH-SY5Y cell line. Exponentially growing cells were maintained in DMEM/F12 medium and differentiation was triggered by the combination of retinoic acid (RA) and BDNF. Both acetylcholinesterase (AChE) and choline acetyltransferase (ChAT) enzymatic activities and immunocontent were determined. For mimicking tau and amyloid-β pathology, RA + BDNF-differentiated cells were challenged with okadaic acid (OA) or soluble oligomers of amyloid-β (AβOs) and neurotoxicity was evaluated. RA + BDNF-induced differentiation resulted in remarkable neuronal morphology alterations characterized by increased neurite density. Enhanced expression and enzymatic activities of cholinergic markers were observed compared to RA-differentiation only. Combination of sublethal doses of AβOs and OA resulted in decreased neurite densities, an in vitro marker of synaptopathy. Challenging RA + BDNF-differentiated SH-SY5Y cells with the combination of sublethal doses of OA and AβO, without causing considerable decrease of cell viability, provides an in vitro model which mimics the early-stage pathophysiology of cholinergic neurons affected by AD.
Collapse
|
6
|
Pezzini F, Bettinetti L, Di Leva F, Bianchi M, Zoratti E, Carrozzo R, Santorelli FM, Delledonne M, Lalowski M, Simonati A. Transcriptomic Profiling Discloses Molecular and Cellular Events Related to Neuronal Differentiation in SH-SY5Y Neuroblastoma Cells. Cell Mol Neurobiol 2017; 37:665-682. [PMID: 27422411 DOI: 10.1007/s10571-016-0403-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
Human SH-SY5Y neuroblastoma cells are widely utilized in in vitro studies to dissect out pathogenetic mechanisms of neurodegenerative disorders. These cells are considered as neuronal precursors and differentiate into more mature neuronal phenotypes under selected growth conditions. In this study, in order to decipher the pathways and cellular processes underlying neuroblastoma cell differentiation in vitro, we performed systematic transcriptomic (RNA-seq) and bioinformatic analysis of SH-SY5Y cells differentiated according to a two-step paradigm: retinoic acid treatment followed by enriched neurobasal medium. Categorization of 1989 differentially expressed genes (DEGs) identified in differentiated cells functionally linked them to changes in cell morphology including remodelling of plasma membrane and cytoskeleton, and neuritogenesis. Seventy-three DEGs were assigned to axonal guidance signalling pathway, and the expression of selected gene products such as neurotrophin receptors, the functionally related SLITRK6, and semaphorins, was validated by immunoblotting. Along with these findings, the differentiated cells exhibited an ability to elongate longer axonal process as assessed by the neuronal cytoskeletal markers biochemical characterization and morphometric evaluation. Recognition of molecular events occurring in differentiated SH-SY5Y cells is critical to accurately interpret the cellular responses to specific stimuli in studies on disease pathogenesis.
Collapse
Affiliation(s)
- Francesco Pezzini
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Laura Bettinetti
- Department of Biotechnologies, University of Verona, Verona, Italy
| | | | - Marzia Bianchi
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Elisa Zoratti
- Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
- Aptuit s.r.l., Verona, Italy
| | - Rosalba Carrozzo
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Filippo M Santorelli
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, IRCCS Stella Maris, Calambrone-Pisa, Italy
| | | | - Maciej Lalowski
- Medicum, Biochemistry/Developmental Biology Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki, Finland.
| | - Alessandro Simonati
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
| |
Collapse
|
7
|
Forster JI, Köglsberger S, Trefois C, Boyd O, Baumuratov AS, Buck L, Balling R, Antony PMA. Characterization of Differentiated SH-SY5Y as Neuronal Screening Model Reveals Increased Oxidative Vulnerability. ACTA ACUST UNITED AC 2016; 21:496-509. [PMID: 26738520 PMCID: PMC4904349 DOI: 10.1177/1087057115625190] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/10/2015] [Indexed: 02/01/2023]
Abstract
The immortalized and proliferative cell line SH-SY5Y is one of the most commonly used cell lines in neuroscience and neuroblastoma research. However, undifferentiated SH-SY5Y cells share few properties with mature neurons. In this study, we present an optimized neuronal differentiation protocol for SH-SY5Y that requires only two work steps and 6 days. After differentiation, the cells present increased levels of ATP and plasma membrane activity but reduced expression of energetic stress response genes. Differentiation results in reduced mitochondrial membrane potential and decreased robustness toward perturbations with 6-hydroxydopamine. We are convinced that the presented differentiation method will leverage genetic and chemical high-throughput screening projects targeting pathways that are involved in the selective vulnerability of neurons with high energetic stress levels.
Collapse
Affiliation(s)
- J I Forster
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - S Köglsberger
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - C Trefois
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - O Boyd
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - A S Baumuratov
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - L Buck
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - R Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - P M A Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| |
Collapse
|
8
|
Pakladok T, Hosseinzadeh Z, Almilaji A, Lebedeva A, Shumilina E, Alesutan I, Lang F. Up-regulation of hERG K⁺ channels by B-RAF. PLoS One 2014; 9:e87457. [PMID: 24475291 PMCID: PMC3903650 DOI: 10.1371/journal.pone.0087457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/26/2013] [Indexed: 11/19/2022] Open
Abstract
Human ether-a-go-go related-gene K⁺ channels (hERG) participate in the regulation of tumor cell proliferation and apoptosis. HERG channel activity is up-regulated by growth factors. Kinases sensitive to growth factor signaling include the serine/threonine protein kinase B-RAF. The present study thus explored whether B-RAF influences hERG channel expression and activity. To this end, hERG channels were expressed in Xenopus oocytes with or without wild-type B-RAF, hERG channel activity was determined utilizing dual-electrode voltage clamp and hERG protein abundance in the cell membrane was analyzed utilizing confocal microscopy as well as chemiluminescence. Moreover, in rhabdomyosarcoma RD cells the effect of B-RAF inhibitor PLX-4720 on hERG-mediated current was quantified by whole-cell patch clamp and hERG cell surface protein abundance by utilizing biotinylation of cell surface proteins as well as flow cytometry. As a result, co-expression of wild-type B-RAF in hERG-expressing Xenopus oocytes significantly increased hERG channel activity and hERG channel protein abundance in the cell membrane. Treatment for 24 hours of B-RAF and hERG-expressing Xenopus oocytes with B-RAF inhibitor PLX-4720 (10 µM) significantly decreased hERG-mediated current and hERG cell surface expression. Similarly, in rhabdomyosarcoma RD cells, treatment for 24 hours with B-RAF inhibitor PLX-4720 significantly decreased hERG cell membrane protein abundance and hERG-mediated current. In conclusion, B-RAF is a powerful regulator of hERG channel activity and cell surface hERG protein abundance.
Collapse
Affiliation(s)
| | | | - Ahmad Almilaji
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Aleksandra Lebedeva
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia
| | | | - Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Lansu K, Gentile S. Potassium channel activation inhibits proliferation of breast cancer cells by activating a senescence program. Cell Death Dis 2013; 4:e652. [PMID: 23744352 PMCID: PMC3698542 DOI: 10.1038/cddis.2013.174] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traditionally the hERG1 potassium channel has been known to have a fundamental role in membrane excitability of several mammalian cells including cardiac myocytes. hERG1 has recently been found to be expressed in non-excitable cancer cells of different histogenesis, but the role of this channel in cancer biology is unknown. Results form recent studies on the effect hERG1 inhibition in some breast cancer cells are controversial as it can lead to apoptosis or protect against cell death. Nevertheless, these data suggest that the hERG1 channel could have an important role in cancer biology. Here we report the effects of hyperstimulation of hERG1 channel in human mammary gland adenocarcinoma-derived cells. Application of the hERG1 activator, the diphenylurea derivative NS1643, inhibits cell proliferation irreversibly. This event is accompanied by a preferential arrest of the cell cycle in G0/G1 phase without the occurrence of apoptotic events. Consequently, cells responded to NS1643 by developing a senescence-like phenotype associated with increased protein levels of the tumor suppressors p21 and p16(INK4a) and by a positive β-galactosidase assay. These data suggest that prolonged stimulation of the hERG1 potassium channel may activate a senescence program and offers a compelling opportunity to develop a potential antiproliferative cancer therapy.
Collapse
Affiliation(s)
- K Lansu
- Molecular Pharmacology and Therapeutics, Loyola University, Chicago, IL, USA
| | | |
Collapse
|
10
|
Sundelacruz S, Levin M, Kaplan DL. Depolarization alters phenotype, maintains plasticity of predifferentiated mesenchymal stem cells. Tissue Eng Part A 2013; 19:1889-908. [PMID: 23738690 DOI: 10.1089/ten.tea.2012.0425.rev] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although adult stem cell transplantation has been implemented as a therapy for tissue repair, it is limited by the availability of functional adult stem cells. A potential approach to generate stem and progenitor cells may be to modulate the differentiated status of somatic cells. Therefore, there is a need for a better understanding of how the differentiated phenotype of mature cells is regulated. We hypothesize that bioelectric signaling plays an important role in the maintenance of the differentiated state, as it is a functional regulator of the differentiation process in various cells and tissues. In this study, we asked whether the mature phenotype of osteoblasts and adipocytes derived from human mesenchymal stem cells (hMSCs) could be altered by modulation of their membrane potential. hMSC-derived osteoblasts and adipocytes were depolarized by treatment with ouabain, a Na(+)/K(+) ATPase inhibitor, or by treatment with high concentrations of extracellular K(+). To characterize the effect of voltage modulation on the differentiated state, the depolarized cells were evaluated for (1) the loss of differentiation markers; (2) the up-regulation of stemness markers and stem properties; and (3) differences in gene expression profiles in response to voltage modulation. hMSC-derived osteoblasts and adipocytes exhibited significant down-regulation of bone and fat tissue markers in response to depolarization, despite the presence of differentiation-inducing soluble factors, suggesting that bioelectric signaling overrides biochemical signaling in the maintenance of cell state. Suppression of the osteoblast or adipocyte phenotype was not accompanied by up-regulation of genes associated with the stem state. Thus, depolarization does not activate the stem cell genetic signature and, therefore, does not induce a full reprogramming event. However, after transdifferentiating the depolarized cells to evaluate for multi-lineage potential, depolarized osteoblasts demonstrated improved ability to achieve correct adipocyte morphology compared with nondepolarized osteoblasts. The present study thus demonstrates that depolarization reduces the differentiated phenotype of hMSC-derived cells and improves their transdifferentiation capacity, but does not restore a stem-like genetic profile. Through global transcript profiling of depolarized osteoblasts, we identified pathways that may mediate the effects of voltage signaling on cell state, which will require a detailed mechanistic inquiry in future studies.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | | | | |
Collapse
|
11
|
Roepke TK, Purtell K, King EC, La Perle KMD, Lerner DJ, Abbott GW. Targeted deletion of Kcne2 causes gastritis cystica profunda and gastric neoplasia. PLoS One 2010; 5:e11451. [PMID: 20625512 PMCID: PMC2897890 DOI: 10.1371/journal.pone.0011451] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 06/13/2010] [Indexed: 01/02/2023] Open
Abstract
Gastric cancer is the second leading cause of cancer death worldwide. Predisposing factors include achlorhydria, Helicobacter pylori infection, oxyntic atrophy and TFF2-expressing metaplasia. In parietal cells, apical potassium channels comprising the KCNQ1 α subunit and the KCNE2 β subunit provide a K+ efflux current to facilitate gastric acid secretion by the apical H+K+ATPase. Accordingly, genetic deletion of murine Kcnq1 or Kcne2 impairs gastric acid secretion. Other evidence has suggested a role for KCNE2 in human gastric cancer cell proliferation, independent of its role in gastric acidification. Here, we demonstrate that 1-year-old Kcne2−/− mice in a pathogen-free environment all exhibit a severe gastric preneoplastic phenotype comprising gastritis cystica profunda, 6-fold increased stomach mass, increased Ki67 and nuclear Cyclin D1 expression, and TFF2- and cytokeratin 7-expressing metaplasia. Some Kcne2−/−mice also exhibited pyloric polypoid adenomas extending into the duodenum, and neoplastic invasion of thin walled vessels in the sub-mucosa. Finally, analysis of human gastric cancer tissue indicated reduced parietal cell KCNE2 expression. Together with previous findings, the results suggest KCNE2 disruption as a possible risk factor for gastric neoplasia.
Collapse
Affiliation(s)
- Torsten K. Roepke
- Department of Pharmacology, Weill Medical College of Cornell University, New York, New York, United States of America
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Kerry Purtell
- Department of Pharmacology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Elizabeth C. King
- Department of Pharmacology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Krista M. D. La Perle
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Daniel J. Lerner
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Geoffrey W. Abbott
- Department of Pharmacology, Weill Medical College of Cornell University, New York, New York, United States of America
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Sundelacruz S, Levin M, Kaplan DL. Role of membrane potential in the regulation of cell proliferation and differentiation. Stem Cell Rev Rep 2009; 5:231-46. [PMID: 19562527 PMCID: PMC10467564 DOI: 10.1007/s12015-009-9080-2] [Citation(s) in RCA: 329] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 06/07/2009] [Indexed: 12/11/2022]
Abstract
Biophysical signaling, an integral regulator of long-term cell behavior in both excitable and non-excitable cell types, offers enormous potential for modulation of important cell functions. Of particular interest to current regenerative medicine efforts, we review several examples that support the functional role of transmembrane potential (V(mem)) in the regulation of proliferation and differentiation. Interestingly, distinct V(mem) controls are found in many cancer cell and precursor cell systems, which are known for their proliferative and differentiation capacities, respectively. Collectively, the data demonstrate that bioelectric properties can serve as markers for cell characterization and can control cell mitotic activity, cell cycle progression, and differentiation. The ability to control cell functions by modulating bioelectric properties such as V(mem) would be an invaluable tool for directing stem cell behavior toward therapeutic goals. Biophysical properties of stem cells have only recently begun to be studied and are thus in need of further characterization. Understanding the molecular and mechanistic basis of biophysical regulation will point the way toward novel ways to rationally direct cell functions, allowing us to capitalize upon the potential of biophysical signaling for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA 02155, USA
| | | | | |
Collapse
|
15
|
Sundelacruz S, Levin M, Kaplan DL. Membrane potential controls adipogenic and osteogenic differentiation of mesenchymal stem cells. PLoS One 2008; 3:e3737. [PMID: 19011685 PMCID: PMC2581599 DOI: 10.1371/journal.pone.0003737] [Citation(s) in RCA: 181] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 10/28/2008] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Control of stem cell behavior is a crucial aspect of developmental biology and regenerative medicine. While the functional role of electrophysiology in stem cell biology is poorly understood, it has become clear that endogenous ion flows represent a powerful set of signals by means of which cell proliferation, differentiation, and migration can be controlled in regeneration and embryonic morphogenesis. METHODOLOGY/PRINCIPAL FINDINGS We examined the membrane potential (V(mem)) changes exhibited by human mesenchymal stem cells (hMSCs) undergoing adipogenic (AD) and osteogenic (OS) differentiation, and uncovered a characteristic hyperpolarization of differentiated cells versus undifferentiated cells. Reversal of the progressive polarization via pharmacological modulation of transmembrane potential revealed that depolarization of hMSCs prevents differentiation. In contrast, treatment with hyperpolarizing reagents upregulated osteogenic markers. CONCLUSIONS/SIGNIFICANCE Taken together, these data suggest that the endogenous hyperpolarization is a functional determinant of hMSC differentiation and is a tractable control point for modulating stem cell function.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Michael Levin
- Biology Department, and Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, United States of America
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
16
|
Guasti L, Crociani O, Redaelli E, Pillozzi S, Polvani S, Masselli M, Mello T, Galli A, Amedei A, Wymore RS, Wanke E, Arcangeli A. Identification of a posttranslational mechanism for the regulation of hERG1 K+ channel expression and hERG1 current density in tumor cells. Mol Cell Biol 2008; 28:5043-60. [PMID: 18559421 PMCID: PMC2519704 DOI: 10.1128/mcb.00304-08] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 03/25/2008] [Accepted: 06/04/2008] [Indexed: 01/19/2023] Open
Abstract
A common feature of tumor cells is the aberrant expression of ion channels on their plasma membrane. The molecular mechanisms regulating ion channel expression in cancer cells are still poorly known. K(+) channels that belong to the human ether-a-go-go-related gene 1 (herg1) family are frequently misexpressed in cancer cells compared to their healthy counterparts. We describe here a posttranslational mechanism for the regulation of hERG1 channel surface expression in cancer cells. This mechanism is based on the activity of hERG1 isoforms containing the USO exon. These isoforms (i) are frequently overexpressed in human cancers, (ii) are retained in the endoplasmic reticulum, and (iii) form heterotetramers with different proteins of the hERG family. (iv) The USO-containing heterotetramers are retained intracellularly and undergo ubiquitin-dependent degradation. This process results in decreased hERG1 current (I(hERG1)) density. We detailed such a mechanism in heterologous systems and confirmed its functioning in tumor cells that endogenously express hERG1 proteins. The silencing of USO-containing hERG1 isoforms induces a higher I(hERG1) density in tumors, an effect that apparently regulates neurite outgrowth in neuroblastoma cells and apoptosis in leukemia cells.
Collapse
Affiliation(s)
- Leonardo Guasti
- Department of Experimental Pathology and Oncology, University of Florence, Viale G. B. Morgagni 50, 50134 Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wasicko MJ, Breitwieser GE, Kim I, Carroll JL. Postnatal development of carotid body glomus cell response to hypoxia. Respir Physiol Neurobiol 2006; 154:356-71. [PMID: 16466972 DOI: 10.1016/j.resp.2006.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 01/10/2006] [Accepted: 01/11/2006] [Indexed: 10/25/2022]
Abstract
This study examines developmental changes in CB glomus cell depolarization, intracellular calcium ([Ca(2+)](i)) and the magnitude of an O(2)-sensitive background ionic conductance that may play roles in the postnatal increase in oxygen sensitivity of glomus cells isolated from rats of 1-3 days and 11-14 days postnatal age. Using fura-2 and perforated patch whole cell recordings, we simultaneously measured [Ca(2+)](i) and membrane potential (E(m)) during normoxia and hypoxia. Resting E(m) in normoxia was similar at both ages. Hypoxia caused a larger E(m) depolarization and correspondingly larger [Ca(2+)](i) response in glomus cells from 11- to 14-day-old rats compared to 1-3-day-old rats. E(m) and [Ca(2+)](i) responses to 40mM K(+) were identical between the two age groups. Under normoxic conditions both age groups had similar background conductances. Under anoxic conditions (at resting membrane potential) background K(+) conductance decreased significantly more in cells from 11- to 14-day-old rats compared to cells from 1- to 3-day-old rats. Glomus cells from newborns therefore have less O(2)-sensitive background K(+) conductance. These results support the hypothesis that postnatal maturation of glomus cell O(2) sensitivity involves developmental regulation of the expression and/or O(2)-sensitivity of background ionic conductances.
Collapse
Affiliation(s)
- M J Wasicko
- Johns Hopkins University School of Medicine, Johns Hopkins Children's Center, Baltimore, MD, United States
| | | | | | | |
Collapse
|
18
|
Biagiotti T, D'Amico M, Marzi I, Di Gennaro P, Arcangeli A, Wanke E, Olivotto M. Cell Renewing in Neuroblastoma: Electrophysiological and Immunocytochemical Characterization of Stem Cells and Derivatives. Stem Cells 2006; 24:443-53. [PMID: 16100002 DOI: 10.1634/stemcells.2004-0264] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We explored the stem cell compartment of the SH-SY5Y neuroblastoma (NB) clone and its development by a novel approach, integrating clonal and immunocytochemical investigations with patch-clamp measurements of ion currents simultaneously expressed on single cells. The currents selected were the triad IHERG, IKDR, INa, normally expressed at varying mutual ratios during development of neural crest stem cells, from which NB derives upon neoplastic transformation. These ratios could be used as electrophysiological clusters of differentiation (ECDs), identifying otherwise indistinguishable stages in maturation. Subcloning procedures allowed the isolation of highly clonogenic substrate-adherent (S-type) cells that proved to be p75- and nestinpositive and were characterized by a nude electrophysiological profile (ECDS0). These cells expressed negligible levels of the triad and manifested the capacity of generating the two following lineages: first, a terminally differentiating, smooth muscular lineage, positive for calponin and smooth muscle actin, whose electrophysiological profile is characterized by a progressive diminution of IHERG, the increase of IKDR and INa, and the acquisition of IKIR (ECDS2); second, a neuronal abortive pathway (NF-68 positive), characterized by a variable expression of IHERG and IKDR and a low expression of INa (ECDNS). This population manifested a vigorous amplification, monopolizing the stem cell compartment at the expense of the smooth muscular lineage to such an extent that neuronal-like (N-type) cells must be continuously removed if the latter are to develop.
Collapse
Affiliation(s)
- Tiziana Biagiotti
- Department of Experimental Pathology and Oncology, University of Forence, Italy
| | | | | | | | | | | | | |
Collapse
|
19
|
Carroll JL, Kim I. Postnatal development of carotid body glomus cell O2 sensitivity. Respir Physiol Neurobiol 2005; 149:201-15. [PMID: 15886071 DOI: 10.1016/j.resp.2005.04.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 04/07/2005] [Accepted: 04/07/2005] [Indexed: 01/17/2023]
Abstract
In mammals, the main sensors of arterial oxygen level are the carotid chemoreceptors, which exhibit low sensitivity to hypoxia at birth and become more sensitive over the first few days or weeks of life. This postnatal increase in hypoxia sensitivity of the arterial chemoreceptors, termed "resetting", remains poorly understood. In the carotid body, hypoxia is transduced by glomus cells, which are secretory sensory neurons that respond to hypoxia at higher P(O2) levels than non-chemoreceptor cell types. Maturation or resetting of carotid body O2 sensitivity potentially involves numerous aspects of the O2 transduction cascade at the glomus cell level, including glomus cell neurotransmitter secretion, neuromodulator function, neurotransmitter receptor expression, glomus cell depolarization in response to hypoxia, [Ca2+]i responses to hypoxia, K+ and Ca2+ channel O2 sensitivity and K+ channel expression. However, although progress has been made in the understanding of carotid body development, the precise mechanisms underlying postnatal maturation of these numerous aspects of chemotransduction remain obscure.
Collapse
Affiliation(s)
- John L Carroll
- University of Arkansas for Medical Sciences College of Medicine, Department of Pediatrics, Pediatric Respiratory Medicine Section, Arkansas Children's Hospital, Little Rock, AR 72202, USA.
| | | |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Garcia-Gil M, Pesi R, Perna S, Allegrini S, Giannecchini M, Camici M, Tozzi MG. 5'-aminoimidazole-4-carboxamide riboside induces apoptosis in human neuroblastoma cells. Neuroscience 2003; 117:811-20. [PMID: 12654334 DOI: 10.1016/s0306-4522(02)00836-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
5'-Aminoimidazole-4-carboxamide riboside (AICA riboside) has been previously shown to be toxic to two neuronal cell models [Neuroreport 11 (2000) 1827]. In this paper we demonstrate that AICA riboside promotes apoptosis in undifferentiated human neuroblastoma cells (SH-SY5Y), inducing a raise in caspase-3 activity. In order to exert its effect on viability, AICA riboside must enter the cells and be phosphorylated to the ribotide, since both a nucleoside transport inhibitor, and an inhibitor of adenosine kinase produce an enhancement of the viability of AICA riboside-treated cells. Short-term incubations (2 h) with AICA riboside result in five-fold increase in the activity of AMP-dependent protein kinase (AMPK). However, the activity of AMPK is not significantly affected at prolonged incubations (48 h), when the apoptotic effect of AICA riboside is evident. The results demonstrate that when the cell line is induced to differentiate both toward a cholinergic phenotype (with retinoic acid) or a noradrenergic phenotype (with phorbol esters), the toxic effect is significantly reduced, and in the case of the noradrenergic phenotype differentiation, the riboside is completely ineffective in promoting apoptosis. This reduction of effect correlates with an overexpression of Bcl-2 during differentiation. AICA riboside, derived from the hydrolysis of the ribotide, an intermediate of purine de novo synthesis, is absent in normal healthy cells; however it may accumulate in those individuals in which an inborn error of purine metabolism causes an increase in the rate of de novo synthesis and/or an overexpression of cytosolic 5'-nucleotidase, that appears to be the enzyme responsible for AICA ribotide hydrolysis. In fact, 5'-nucleotidase activity has been shown to increase in patients affected by Lesch-Nyhan syndrome in which both acceleration of de novo synthesis and accumulation of AICA ribotide has been described, and also in other neurological disorders of unknown etiology. Our results raise the intriguing clue that the neurotoxic effect of AICA riboside on the developing brain might contribute to the neurological manifestations of syndromes related to purine dismetabolisms.
Collapse
Affiliation(s)
- M Garcia-Gil
- Dipartimento di Fisiologia e Biochimica, Università di Pisa, Via S. Maria, 55 56100, Pisa, Italy.
| | | | | | | | | | | | | |
Collapse
|
23
|
Crociani O, Guasti L, Balzi M, Becchetti A, Wanke E, Olivotto M, Wymore RS, Arcangeli A. Cell cycle-dependent expression of HERG1 and HERG1B isoforms in tumor cells. J Biol Chem 2003; 278:2947-55. [PMID: 12431979 DOI: 10.1074/jbc.m210789200] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The role of K(+) channel activity during cell cycle progression has become a research topic of considerable interest. Blocking of K(+) channels inhibits the proliferation of many cell types, although the mechanism of this inhibition is unclear. There is speculation that K(+) channels differentially regulate the electrical potential of the plasma membrane (V(m)) during proliferation. We have demonstrated that in tumor cells the value of V(m) is clamped to rather depolarized values by K(+) channels belonging to the HERG family. We report here that tumor cell lines preferentially express the herg1 gene and a truncated, N-deleted form that corresponds to herg1b. This alternative transcript is also expressed in human primary acute myeloid leukemias. Both HERG1 and HERG1B proteins are expressed on the plasma membrane of tumor cells and can form heterotetramers. The expression of HERG protein isoforms is strongly cell cycle-dependent, accounting for variations in HERG currents along the mitotic cycle. Moreover, the blocking of HERG channels dramatically impairs cell growth of HERG-bearing tumor cells. These results suggest that modulated expression of different K(+) channels is the molecular basis of a novel mechanism regulating neoplastic cell proliferation.
Collapse
Affiliation(s)
- Olivia Crociani
- Department of Experimental Pathology and Oncology, University of Firenze, Viale G. B. Morgagni 50, 50134 Firenze, Italy
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Witchel HJ, Milnes JT, Mitcheson JS, Hancox JC. Troubleshooting problems with in vitro screening of drugs for QT interval prolongation using HERG K+ channels expressed in mammalian cell lines and Xenopus oocytes. J Pharmacol Toxicol Methods 2002; 48:65-80. [PMID: 14565563 DOI: 10.1016/s1056-8719(03)00041-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The majority of drugs associated with QT interval prolongation share an ability to inhibit ionic currents passed by HERG potassium channels. One method of screening new chemical entities (NCEs) for QT prolonging potential is therefore to use heterologous systems expressing HERG channels. Such systems are also of value in the understanding of the function, kinetics, sorting, pharmacological sensitivities, and important molecular determinants of the HERG potassium channel. The methods for incorporating the HERG potassium channel into cells and measuring the consequent current are a mixture of techniques that are standard (for heterologous expression of most ion channels) and individualised to HERG. This review presents a selection of the most commonly used methods for examining heterologous HERG currents, as well as introducing some of the technical problems that may be encountered and their solutions. In mammalian cell lines, problems such as fragile membranes, high leak currents, inability to form a gigaseal, diminished HERG current, endogenous transient outward current, altered kinetics, and even occasional run down can interfere with measurements. In Xenopus oocytes, endogenous chloride currents, insufficient superfusate flow, diminished HERG current and HERG current 'run up' may create difficulties.
Collapse
Affiliation(s)
- Harry J Witchel
- Cardiovascular Research Laboratories, Department of Physiology, School of Medical Sciences, University Walk, Bristol BS8 1TD, UK.
| | | | | | | |
Collapse
|
25
|
Smith GAM, Tsui HW, Newell EW, Jiang X, Zhu XP, Tsui FWL, Schlichter LC. Functional up-regulation of HERG K+ channels in neoplastic hematopoietic cells. J Biol Chem 2002; 277:18528-34. [PMID: 11893742 DOI: 10.1074/jbc.m200592200] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Kv1.3 channels regulate proliferation of normal lymphocytes, but the role of voltage-gated potassium channels in transformed hematopoietic cells is not known. We examined transcripts for Kv1.3, h-erg, h-eag, and BEC1 genes in primary lymphocytes and leukemias and in several hematopoietic cell lines. Surprisingly, BEC1, formerly thought to be brain-specific, was present in all the primary leukemias examined, in resting peripheral blood lymphocytes, and in proliferating activated tonsillar cells, lymphocytes from Sjögren's patients, and Epstein-Barr virus-transformed B-cells. Only h-erg mRNA was up-regulated in the cancer cells, but this was not due to proliferation per se, because it was not elevated in any of the proliferating noncancerous lymphocyte types examined. Nor did h-erg transcript levels correlate with the B-cell subset, because it was elevated in immature neoplastic B-CLL cells (CD5(+)) and in a CD5(-) Burkitt's lymphoma cell line (Raji) but not in Sjögren's syndrome cells (enriched in CD5(+) B-cells) or Epstein-Barr virus-transformed B-cells, which are mature CD5(-) B-cells. The protein and whole cell current levels roughly corresponded with the amount of mRNA expressed in three hematopoietic cell lines: CEM (an acute lymphoblastic leukemic line), K562 (a chronic myelogenous leukemic line), and U937 (an acute promyelocytic leukemic line). The selective HERG channel blocker, E-4031, reduced proliferation of CEM, U937, and K562 cells, and this appears to be the first direct evidence of a functional role for the HERG current in cancer cells. Selective up-regulation of h-erg appears to occur in neoplastic hematopoietic cells, thus providing a marker and potential therapeutic target.
Collapse
Affiliation(s)
- Garth A M Smith
- Division of Cellular and Molecular Biology, Toronto Western Research Institute, Toronto, Ontario M5T 2S8, Canada
| | | | | | | | | | | | | |
Collapse
|
26
|
Hofmann G, Bernabei PA, Crociani O, Cherubini A, Guasti L, Pillozzi S, Lastraioli E, Polvani S, Bartolozzi B, Solazzo V, Gragnani L, Defilippi P, Rosati B, Wanke E, Olivotto M, Arcangeli A. HERG K+ channels activation during beta(1) integrin-mediated adhesion to fibronectin induces an up-regulation of alpha(v)beta(3) integrin in the preosteoclastic leukemia cell line FLG 29.1. J Biol Chem 2001; 276:4923-31. [PMID: 11080495 DOI: 10.1074/jbc.m005682200] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin receptors have been demonstrated to mediate either "inside-to-out" and "outside-to-in" signals, and by this way are capable of regulating many cellular functions, such as cell growth and differentiation, cell migration, and activation. Among the various integrin-centered signaling pathways discovered so far, we demonstrated that the modulation of the electrical potential of the plasma membrane (V(REST)) is an early integrin-mediated signal, which is related to neurite emission in neuroblastoma cells. This modulation is sustained by the activation of HERG K(+) channels, encoded by the ether-à-go-go-related gene (herg). The involvement of integrin-mediated signaling is being discovered in the hemopoietic system: in particular, osteoclasts are generated as well as induced to differentiate by interaction of osteoclast progenitors with the stromal cells, through the involvement of integrin receptors. We studied the effects of cell interaction with the extracellular matrix protein fibronectin (FN) in a human leukemic preosteoclastic cell line (FLG 29.1 cells), which has been demonstrated to express HERG currents. We report here that FLG 29.1 cells indeed adhere to purified FN through integrin receptors, and that this adhesion induces an osteoclast phenotype in these cells, as evidenced by the appearance of tartrate-resistant acid phosphatase, as well as by the increased expression of CD51/alpha(v)beta(3) integrin and calcitonin receptor. An early activation of HERG current (I(HERG)), without any increase in herg RNA or modifications of HERG protein was also observed in FN-adhering cells. This activation is apparently sustained by the beta(1) integrin subunit activation, through the involvement of a pertussis-toxin sensitive G(i) protein, and appears to be a determinant signal for the up-regulation of alpha(v)beta(3) integrin, as well as for the increased expression of calcitonin receptor.
Collapse
Affiliation(s)
- G Hofmann
- Department of Experimental Pathology and Oncology, University of Firenze, Viale G. B. Morgagni, 50, 50134 Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abbott GW, Butler MH, Bendahhou S, Dalakas MC, Ptacek LJ, Goldstein SA. MiRP2 forms potassium channels in skeletal muscle with Kv3.4 and is associated with periodic paralysis. Cell 2001; 104:217-31. [PMID: 11207363 DOI: 10.1016/s0092-8674(01)00207-0] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The subthreshold, voltage-gated potassium channel of skeletal muscle is shown to contain MinK-related peptide 2 (MiRP2) and the pore-forming subunit Kv3.4. MiRP2-Kv3.4 channels differ from Kv3.4 channels in unitary conductance, voltage-dependent activation, recovery from inactivation, steady-state open probability, and block by a peptide toxin. Thus, MiRP2-Kv3.4 channels set resting membrane potential (RMP) and do not produce afterhyperpolarization or cumulative inactivation to limit action potential frequency. A missense mutation is identified in the gene for MiRP2 (KCNE3) in two families with periodic paralysis and found to segregate with the disease. Mutant MiRP2-Kv3.4 complexes exhibit reduced current density and diminished capacity to set RMP. Thus, MiRP2 operates with a classical potassium channel subunit to govern skeletal muscle function and pathophysiology.
Collapse
Affiliation(s)
- G W Abbott
- Departments of Pediatrics and Cellular, Molecular Physiology, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, CT 06536, USA
| | | | | | | | | | | |
Collapse
|
28
|
Encinas M, Iglesias M, Liu Y, Wang H, Muhaisen A, Ceña V, Gallego C, Comella JX. Sequential treatment of SH-SY5Y cells with retinoic acid and brain-derived neurotrophic factor gives rise to fully differentiated, neurotrophic factor-dependent, human neuron-like cells. J Neurochem 2000; 75:991-1003. [PMID: 10936180 DOI: 10.1046/j.1471-4159.2000.0750991.x] [Citation(s) in RCA: 575] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A rapid and simple procedure is presented to obtain nearly pure populations of human neuron-like cells from the SH-SY5Y neuroblastoma cell line. Sequential exposure of SH-SY5Y cells to retinoic acid and brain-derived neurotrophic factor in serum-free medium yields homogeneous populations of cells with neuronal morphology, avoiding the presence of other neural crest derivatives that would normally arise from those cells. Cells are withdrawn from the cell cycle, as shown by 5-bromo-2'-deoxyuridine uptake and retinoblastoma hypophosphorylation. Cell survival is dependent on the continuous presence of brain-derived neurotrophic factor, and removal of this neurotrophin causes apoptotic cell death accompanied by an attempt to reenter the cell cycle. Differentiated cells express neuronal markers, including neurofilaments, neuron-specific enolase, and growth-associated protein-43 as well as neuronal polarity markers such as tau and microtubule-associated protein 2. Moreover, differentiated cultures do not contain glial cells, as could be evidenced after the negative staining for glial fibrillary acidic protein. In conclusion, the protocol presented herein yields homogeneous populations of human neuronal differentiated cells that present many of the characteristics of primary cultures of neurons. This model may be useful to perform large-scale biochemical and molecular studies due to its susceptibility to genetic manipulation and the availability of an unlimited amount of cells.
Collapse
Affiliation(s)
- M Encinas
- Grup de Neurobiologia Molecular, Department de Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida, Lleida, Catalonia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Rosenbaum C, Kamleiter M, Grafe P, Kluwe L, Mautner V, Müller HW, Hanemann CO. Enhanced proliferation and potassium conductance of Schwann cells isolated from NF2 schwannomas can be reduced by quinidine. Neurobiol Dis 2000; 7:483-91. [PMID: 10964617 DOI: 10.1006/nbdi.2000.0307] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurofibromatosis type 2 (NF2) is an autosomal dominant disease that is characterized mainly by schwannomas, as well as menigiomas and gliomas. The NF2 gene product merlin/schwannomin acts as a tumor suppressor. Schwann cells derived from NF2 schwannomas showed an enhanced proliferation rate, and electrophysological studies revealed larger K(+) outward currents as compared with controls. Schwann cells isolated from schwannomas of NF2 patients or multiorgan donors were treated with different concentrations of the K(+) current blockers quinidine, tetraethylammonium chloride, and 4-aminopyridine and K(+) outward currents and proliferation rates of these cells were compared. K(+) outward currents of both cell types can be blocked by quinidine. Importantly, treatment with quinidine reduces proliferation of NF2 Schwann cells in a concentration dependent manner but did not reduce proliferation of normal Schwann cells. Therefore, the use of quinidine or quinidine-like components would possibly provide a novel adjuvant therapeutic option for NF2 patients to slow down or freeze growth of schwannomas.
Collapse
Affiliation(s)
- C Rosenbaum
- Department of Neurology, Heinrich-Heine University, Duesseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|