1
|
Navarro-Pérez M, Capera J, Benavente-Garcia A, Cassinelli S, Colomer-Molera M, Felipe A. Kv1.3 in the spotlight for treating immune diseases. Expert Opin Ther Targets 2024; 28:67-82. [PMID: 38316438 DOI: 10.1080/14728222.2024.2315021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Kv1.3 is the main voltage-gated potassium channel of leukocytes from both the innate and adaptive immune systems. Channel function is required for common processes such as Ca2+ signaling but also for cell-specific events. In this context, alterations in Kv1.3 are associated with multiple immune disorders. Excessive channel activity correlates with numerous autoimmune diseases, while reduced currents result in increased cancer prevalence and immunodeficiencies. AREAS COVERED This review offers a general view of the role of Kv1.3 in every type of leukocyte. Moreover, diseases stemming from dysregulations of the channel are detailed, as well as current advances in their therapeutic research. EXPERT OPINION Kv1.3 arises as a potential immune target in a variety of diseases. Several lines of research focused on channel modulation have yielded positive results. However, among the great variety of specific channel blockers, only one has reached clinical trials. Future investigations should focus on developing simpler administration routes for channel inhibitors to facilitate their entrance into clinical trials. Prospective Kv1.3-based treatments will ensure powerful therapies while minimizing undesired side effects.
Collapse
Affiliation(s)
- María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anna Benavente-Garcia
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Magalí Colomer-Molera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Cheng F, Tang YF, Cao Y, Peng SQ, Zhu XR, Sun Y, Wang SH, Wang B, Lu YM. KCNAB2 overexpression inhibits human non-small-cell lung cancer cell growth in vitro and in vivo. Cell Death Discov 2023; 9:382. [PMID: 37852974 PMCID: PMC10584983 DOI: 10.1038/s41420-023-01679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases. NSCLC patients often have poor prognosis demanding urgent identification of novel biomarkers and potential therapeutic targets. KCNAB2 (regulatory beta subunit2 of voltage-gated potassium channel), encoding aldosterone reductase, plays a pivotal role in regulating potassium channel activity. In this research, we tested the expression of KCNAB2 as well as its potential functions in human NSCLC. Bioinformatics analysis shows that expression of KCNAB2 mRNA is significantly downregulated in human NSCLC, correlating with poor overall survival. In addition, decreased KCNAB2 expression was detected in different NSCLC cell lines and local human NSCLC tissues. Exogenous overexpression of KCNAB2 potently suppressed growth, proliferation and motility of established human NSCLC cells and promoted NSCLC cells apoptosis. In contrast, CRISPR/Cas9-induced KCNAB2 knockout further promoted the malignant biological behaviors of NSCLC cells. Protein chip analysis in the KCNAB2-overexpressed NSCLC cells revealed that KCNAB2 plays a possible role in AKT-mTOR cascade activation. Indeed, AKT-mTOR signaling activation was potently inhibited following KCNAB2 overexpression in NSCLC cells. It was however augmented by KCNAB2 knockout. In vivo, the growth of subcutaneous KCNAB2-overexpressed A549 xenografts was significantly inhibited. Collectively, KCNAB2 could be a novel effective gene for prognosis prediction of NSCLC. Targeting KCNAB2 may lead to the development of advanced therapies.
Collapse
Affiliation(s)
- Feng Cheng
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Yu-Fei Tang
- Department of Soochow Medical college, Soochow University, Suzhou, China
| | - Yang Cao
- Department of Respiratory, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Shi-Qing Peng
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Xiao-Ren Zhu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yue Sun
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Shu-Hang Wang
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Bin Wang
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
| | - Yi-Min Lu
- Department of Respiratory, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
3
|
Dupuy M, Gueguinou M, Potier-Cartereau M, Lézot F, Papin M, Chantôme A, Rédini F, Vandier C, Verrecchia F. SK Ca- and Kv1-type potassium channels and cancer: Promising therapeutic targets? Biochem Pharmacol 2023; 216:115774. [PMID: 37678626 DOI: 10.1016/j.bcp.2023.115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Ion channels are transmembrane structures that allow the passage of ions across cell membranes such as the plasma membrane or the membranes of various organelles like the nucleus, endoplasmic reticulum, Golgi apparatus or mitochondria. Aberrant expression of various ion channels has been demonstrated in several tumor cells, leading to the promotion of key functions in tumor development, such as cell proliferation, resistance to apoptosis, angiogenesis, invasion and metastasis. The link between ion channels and these key biological functions that promote tumor development has led to the classification of cancers as oncochannelopathies. Among all ion channels, the most varied and numerous, forming the largest family, are the potassium channels, with over 70 genes encoding them in humans. In this context, this review will provide a non-exhaustive overview of the role of plasma membrane potassium channels in cancer, describing 1) the nomenclature and structure of potassium channels, 2) the role of these channels in the control of biological functions that promotes tumor development such as proliferation, migration and cell death, and 3) the role of two particular classes of potassium channels, the SKCa- and Kv1- type potassium channels in cancer progression.
Collapse
Affiliation(s)
- Maryne Dupuy
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| | | | | | - Frédéric Lézot
- Sorbonne University, INSERM UMR933, Hôpital Trousseau (AP-HP), Paris F-75012, France
| | - Marion Papin
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | | | - Françoise Rédini
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France
| | | | - Franck Verrecchia
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| |
Collapse
|
4
|
Zayas-Arrabal J, Alquiza A, Rodríguez-de-Yurre A, Echeazarra L, Fernández-López V, Gallego M, Casis O. Kv1.3 Channel Blockade Improves Inflammatory Profile, Reduces Cardiac Electrical Remodeling, and Prevents Arrhythmia in Type 2 Diabetic Rats. Cardiovasc Drugs Ther 2023; 37:63-73. [PMID: 34623540 PMCID: PMC9834174 DOI: 10.1007/s10557-021-07264-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 01/16/2023]
Abstract
PURPOSE Kv1.3 channel regulates the activity of lymphocytes, macrophages, or adipose tissue and its blockade reduces inflammatory cytokine secretion and improves insulin sensitivity in animals with metabolic syndrome and in genetically obese mice. Thus, Kv1.3 blockade could be a strategy for the treatment of type 2 diabetes. Elevated circulating levels of TNFα and IL-1b mediate the higher susceptibility to cardiac arrhythmia in type 2 diabetic rats. We hypothesized that Kv1.3 channel blockade with the psoralen PAP1 could have immunomodulatory properties that prevent QTc prolongation and reduce the risk of arrhythmia in type 2 diabetic rats. METHODS Type 2 diabetes was induced to Sprague-Dawley rats by high-fat diet and streptozotocin injection. Diabetic animals were untreated, treated with metformin, or treated with PAP1 for 4 weeks. Plasma glucose, insulin, cholesterol, triglycerides, and cytokine levels were measured using commercial kits. ECG were recorded weekly, and an arrhythmia-inducing protocol was performed at the end of the experimental period. Action potentials were recorded in isolated ventricular cardiomyocytes. RESULTS In diabetic animals, PAP1 normalized glycaemia, insulin resistance, adiposity, and lipid profile. In addition, PAP1 prevented the diabetes-induced repolarization defects through reducing the secretion of the inflammatory cytokines IL-10, IL-12p70, GM-CSF, IFNγ, and TNFα. Moreover, compared to diabetic untreated and metformin-treated animals, those treated with PAP1 had the lowest risk of developing the life-threatening arrhythmia Torsade de Pointes under cardiac challenge. CONCLUSION Kv1.3 inhibition improves diabetes and diabetes-associated low-grade inflammation and cardiac electrical remodeling, resulting in more protection against cardiac arrhythmia compared to metformin.
Collapse
Affiliation(s)
- Julián Zayas-Arrabal
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Amaia Alquiza
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Ainhoa Rodríguez-de-Yurre
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Leyre Echeazarra
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Víctor Fernández-López
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Mónica Gallego
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Oscar Casis
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain.
| |
Collapse
|
5
|
Boyle Y, Johns TG, Fletcher EV. Potassium Ion Channels in Malignant Central Nervous System Cancers. Cancers (Basel) 2022; 14:cancers14194767. [PMID: 36230692 PMCID: PMC9563970 DOI: 10.3390/cancers14194767] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Malignant central nervous system (CNS) cancers are among the most difficult to treat, with low rates of survival and a high likelihood of recurrence. This is primarily due to their location within the CNS, hindering adequate drug delivery and tumour access via surgery. Furthermore, CNS cancer cells are highly plastic, an adaptive property that enables them to bypass targeted treatment strategies and develop drug resistance. Potassium ion channels have long been implicated in the progression of many cancers due to their integral role in several hallmarks of the disease. Here, we will explore this relationship further, with a focus on malignant CNS cancers, including high-grade glioma (HGG). HGG is the most lethal form of primary brain tumour in adults, with the majority of patient mortality attributed to drug-resistant secondary tumours. Hence, targeting proteins that are integral to cellular plasticity could reduce tumour recurrence, improving survival. This review summarises the role of potassium ion channels in malignant CNS cancers, specifically how they contribute to proliferation, invasion, metastasis, angiogenesis, and plasticity. We will also explore how specific modulation of these proteins may provide a novel way to overcome drug resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Yasmin Boyle
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
- Correspondence:
| | - Terrance G. Johns
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| |
Collapse
|
6
|
Zúñiga L, Cayo A, González W, Vilos C, Zúñiga R. Potassium Channels as a Target for Cancer Therapy: Current Perspectives. Onco Targets Ther 2022; 15:783-797. [PMID: 35899081 PMCID: PMC9309325 DOI: 10.2147/ott.s326614] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/08/2022] [Indexed: 12/18/2022] Open
Abstract
Potassium (K+) channels are highly regulated membrane proteins that control the potassium ion flux and respond to different cellular stimuli. These ion channels are grouped into three major families, Kv (voltage-gated K+ channel), Kir (inwardly rectifying K+ channel) and K2P (two-pore K+ channels), according to the structure, to mediate the K+ currents. In cancer, alterations in K+ channel function can promote the acquisition of the so-called hallmarks of cancer – cell proliferation, resistance to apoptosis, metabolic changes, angiogenesis, and migratory capabilities – emerging as targets for the development of new therapeutic drugs. In this review, we focus our attention on the different K+ channels associated with the most relevant and prevalent cancer types. We summarize our knowledge about the potassium channels structure and function, their cancer dysregulated expression and discuss the K+ channels modulator and the strategies for designing new drugs.
Collapse
Affiliation(s)
- Leandro Zúñiga
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Angel Cayo
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca, Chile
| | - Cristian Vilos
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile.,Laboratory of Nanomedicine and Targeted Delivery, School of Medicine, Universidad de Talca, Talca, 3460000, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| | - Rafael Zúñiga
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| |
Collapse
|
7
|
Li P, Kurata Y, Taufiq F, Kuwabara M, Ninomiya H, Higaki K, Tsuneto M, Shirayoshi Y, Lanaspa MA, Hisatome I. Kv1.5 channel mediates monosodium urate-induced activation of NLRP3 inflammasome in macrophages and arrhythmogenic effects of urate on cardiomyocytes. Mol Biol Rep 2022; 49:5939-5952. [PMID: 35368226 PMCID: PMC9270276 DOI: 10.1007/s11033-022-07378-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/15/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Gout is usually found in patients with atrial fibrillation (AF). K+ efflux is a common trigger of NLRP3 inflammasome activation which is involved in the pathogenesis of AF. We investigated the role of the K+ channel Kv1.5 in monosodium urate crystal (MSU)-induced activation of the NLRP3 inflammasome and electrical remodeling in mouse and human macrophages J774.1 and THP-1, and mouse atrial myocytes HL-1. METHODS AND RESULTS Macrophages, primed with lipopolysaccharide (LPS), were stimulated by MSU. HL-1 cells were incubated with the conditioned medium (CM) from MSU-stimulated macrophages. Western blot, ELISA and patch clamp were used. MSU induced caspase-1 expression in LPS-primed J774.1 cells and IL-1β secretion, suggesting NLRP3 inflammasome activation. A selective Kv1.5 inhibitor, diphenyl phosphine oxide-1 (DPO-1), and siRNAs against Kv1.5 suppressed the levels of caspase-1 and IL-1β. MSU reduced intracellular K+ concentration which was prevented by DPO-1 and siRNAs against Kv1.5. MSU increased expression of Hsp70, and Kv1.5 on the plasma membrane. siRNAs against Hsp70 were suppressed but heat shock increased the expression of Hsp70, caspase-1, IL-1β, and Kv1.5 in MSU-stimulated J774.1 cells. The CM from MSU-stimulated macrophages enhanced the expression of caspase-1, IL-1β and Kv1.5 with increased Kv1.5-mediated currents that shortened action potential duration in HL-1 cells. These responses were abolished by DPO-1 and a siRNA against Kv1.5. CONCLUSIONS Kv1.5 regulates MSU-induced activation of NLRP3 inflammasome in macrophages. MSUrelated activation of NLRP3 inflammasome and electrical remodeling in HL-1 cells are via macrophages. Kv1.5 may have therapeutic value for diseases related to gout-induced activation of the NLRP3 inflammsome, including AF.
Collapse
Affiliation(s)
- Peili Li
- Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, 36-1, Nishimachi, Yonago, Tottori, 683-8504, Japan.
| | - Yasutaka Kurata
- Department of Physiology II, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Fikri Taufiq
- Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, 36-1, Nishimachi, Yonago, Tottori, 683-8504, Japan
| | - Masanari Kuwabara
- Intensive Care Unit and Department of Cardiology, Toranomon Hospital, Tokyo, 105-8470, Japan
| | - Haruaki Ninomiya
- Department of Biological Regulation, Tottori University, Yonago, 683-8504, Japan
| | - Katsumi Higaki
- Research Center for Bioscience and Technology, Tottori University, Yonago, 683-8504, Japan
| | - Motokazu Tsuneto
- Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, 36-1, Nishimachi, Yonago, Tottori, 683-8504, Japan
| | - Yasuaki Shirayoshi
- Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, 36-1, Nishimachi, Yonago, Tottori, 683-8504, Japan
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Ichiro Hisatome
- Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, 36-1, Nishimachi, Yonago, Tottori, 683-8504, Japan
| |
Collapse
|
8
|
Borrego J, Feher A, Jost N, Panyi G, Varga Z, Papp F. Peptide Inhibitors of Kv1.5: An Option for the Treatment of Atrial Fibrillation. Pharmaceuticals (Basel) 2021; 14:1303. [PMID: 34959701 PMCID: PMC8704205 DOI: 10.3390/ph14121303] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
The human voltage gated potassium channel Kv1.5 that conducts the IKur current is a key determinant of the atrial action potential. Its mutations have been linked to hereditary forms of atrial fibrillation (AF), and the channel is an attractive target for the management of AF. The development of IKur blockers to treat AF resulted in small molecule Kv1.5 inhibitors. The selectivity of the blocker for the target channel plays an important role in the potential therapeutic application of the drug candidate: the higher the selectivity, the lower the risk of side effects. In this respect, small molecule inhibitors of Kv1.5 are compromised due to their limited selectivity. A wide range of peptide toxins from venomous animals are targeting ion channels, including mammalian channels. These peptides usually have a much larger interacting surface with the ion channel compared to small molecule inhibitors and thus, generally confer higher selectivity to the peptide blockers. We found two peptides in the literature, which inhibited IKur: Ts6 and Osu1. Their affinity and selectivity for Kv1.5 can be improved by rational drug design in which their amino acid sequences could be modified in a targeted way guided by in silico docking experiments.
Collapse
Affiliation(s)
- Jesús Borrego
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Adam Feher
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary;
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Ferenc Papp
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| |
Collapse
|
9
|
Capera J, Pérez-Verdaguer M, Navarro-Pérez M, Felipe A. Kv1.3 Controls Mitochondrial Dynamics during Cell Cycle Progression. Cancers (Basel) 2021; 13:cancers13174457. [PMID: 34503267 PMCID: PMC8431373 DOI: 10.3390/cancers13174457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Voltage-dependent potassium channels control the proliferation of mammalian cells. In addition, mitochondria physiology is highly dynamic during the cell cycle. The aim of this work was to investigate whether the Kv1.3 channel participates in the mitochondrial control of cell cycle progression. Our data confirmed that Kv1.3 facilitates the proliferation of preadipocytes through the control of mitochondrial dynamics. In addition, adipogenesis was also dependent on Kv1.3 expression. We shed light on the role of Kv1.3 in mitochondria and adipose tissue metabolism, contributing further to the control of cell proliferation by Kv1.3. Abstract The voltage-gated potassium channel Kv1.3 is a potential therapeutic target for obesity and diabetes. The genetic ablation and pharmacological inhibition of Kv1.3 lead to a lean phenotype in rodents. The mechanism of regulation of body weight and energy homeostasis involves Kv1.3 expression in different organs, including white and brown adipose tissues. Here, we show that Kv1.3 promotes the proliferation of preadipocytes through the control of mitochondrial dynamics. Kv1.3 is expressed in mitochondria exhibiting high affinity for the perinuclear population. The mitochondrial network is highly dynamic during the cell cycle, showing continuous fusion-fission events. The formation of a hyperfused mitochondrial network at the G1/S phase of the cell cycle is dependent on Kv1.3 expression. Our results demonstrate that Kv1.3 promotes preadipocyte proliferation and differentiation by controlling mitochondrial membrane potential and mitochondrial dynamics at the G1 phase of the cell cycle.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Mireia Pérez-Verdaguer
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
- Correspondence:
| |
Collapse
|
10
|
Potassium and Chloride Ion Channels in Cancer: A Novel Paradigm for Cancer Therapeutics. Rev Physiol Biochem Pharmacol 2021; 183:135-155. [PMID: 34291318 DOI: 10.1007/112_2021_62] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cancer is a collection of diseases caused by specific changes at the genomic level that support cell proliferation indefinitely. Traditionally, ion channels are known to control a variety of cellular processes including electrical signal generation and transmission, secretion, and contraction by controlling ionic gradients. However, recent studies had brought to light important facts on ion channels in cancer biology.In this review we discuss the mechanism linking potassium or chloride ion channel activity to biochemical pathways controlling proliferation in cancer cells and the potential advantages of targeting ion channels as an anticancer therapeutic option.
Collapse
|
11
|
Gubič Š, Hendrickx LA, Toplak Ž, Sterle M, Peigneur S, Tomašič T, Pardo LA, Tytgat J, Zega A, Mašič LP. Discovery of K V 1.3 ion channel inhibitors: Medicinal chemistry approaches and challenges. Med Res Rev 2021; 41:2423-2473. [PMID: 33932253 PMCID: PMC8252768 DOI: 10.1002/med.21800] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/03/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
The KV 1.3 voltage-gated potassium ion channel is involved in many physiological processes both at the plasma membrane and in the mitochondria, chiefly in the immune and nervous systems. Therapeutic targeting KV 1.3 with specific peptides and small molecule inhibitors shows great potential for treating cancers and autoimmune diseases, such as multiple sclerosis, type I diabetes mellitus, psoriasis, contact dermatitis, rheumatoid arthritis, and myasthenia gravis. However, no KV 1.3-targeted compounds have been approved for therapeutic use to date. This review focuses on the presentation of approaches for discovering new KV 1.3 peptide and small-molecule inhibitors, and strategies to improve the selectivity of active compounds toward KV 1.3. Selectivity of dalatazide (ShK-186), a synthetic derivate of the sea anemone toxin ShK, was achieved by chemical modification and has successfully reached clinical trials as a potential therapeutic for treating autoimmune diseases. Other peptides and small-molecule inhibitors are critically evaluated for their lead-like characteristics and potential for progression into clinical development. Some small-molecule inhibitors with well-defined structure-activity relationships have been optimized for selective delivery to mitochondria, and these offer therapeutic potential for the treatment of cancers. This overview of KV 1.3 inhibitors and methodologies is designed to provide a good starting point for drug discovery to identify novel effective KV 1.3 modulators against this target in the future.
Collapse
Affiliation(s)
- Špela Gubič
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Louise A. Hendrickx
- Toxicology and PharmacologyUniversity of Leuven, Campus GasthuisbergLeuvenBelgium
| | - Žan Toplak
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Maša Sterle
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Steve Peigneur
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | | | - Luis A. Pardo
- AG OncophysiologyMax‐Planck Institute for Experimental MedicineGöttingenGermany
| | - Jan Tytgat
- Toxicology and PharmacologyUniversity of Leuven, Campus GasthuisbergLeuvenBelgium
| | - Anamarija Zega
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | | |
Collapse
|
12
|
Capera J, Pérez-Verdaguer M, Peruzzo R, Navarro-Pérez M, Martínez-Pinna J, Alberola-Die A, Morales A, Leanza L, Szabó I, Felipe A. A novel mitochondrial Kv1.3-caveolin axis controls cell survival and apoptosis. eLife 2021; 10:e69099. [PMID: 34196606 PMCID: PMC8248986 DOI: 10.7554/elife.69099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/22/2021] [Indexed: 12/16/2022] Open
Abstract
The voltage-gated potassium channel Kv1.3 plays an apparent dual physiological role by participating in activation and proliferation of leukocytes as well as promoting apoptosis in several types of tumor cells. Therefore, Kv1.3 is considered a potential pharmacological target for immunodeficiency and cancer. Different cellular locations of Kv1.3, at the plasma membrane or the mitochondria, could be responsible for such duality. While plasma membrane Kv1.3 facilitates proliferation, the mitochondrial channel modulates apoptotic signaling. Several molecular determinants of Kv1.3 drive the channel to the cell surface, but no information is available about its mitochondrial targeting. Caveolins, which are able to modulate cell survival, participate in the plasma membrane targeting of Kv1.3. The channel, via a caveolin-binding domain (CDB), associates with caveolin 1 (Cav1), which localizes Kv1.3 to lipid raft membrane microdomains. The aim of our study was to understand the role of such interactions not only for channel targeting but also for cell survival in mammalian cells. By using a caveolin association-deficient channel (Kv1.3 CDBless), we demonstrate here that while the Kv1.3-Cav1 interaction is responsible for the channel localization in the plasma membrane, a lack of such interaction accumulates Kv1.3 in the mitochondria. Kv1.3 CDBless severely affects mitochondrial physiology and cell survival, indicating that a functional link of Kv1.3 with Cav1 within the mitochondria modulates the pro-apoptotic effects of the channel. Therefore, the balance exerted by these two complementary mechanisms fine-tune the physiological role of Kv1.3 during cell survival or apoptosis. Our data highlight an unexpected role for the mitochondrial caveolin-Kv1.3 axis during cell survival and apoptosis.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| | - Mireia Pérez-Verdaguer
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| | | | - María Navarro-Pérez
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| | - Juan Martínez-Pinna
- Dept de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicanteSpain
| | - Armando Alberola-Die
- Dept de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicanteSpain
| | - Andrés Morales
- Dept de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicanteSpain
| | - Luigi Leanza
- Department of Biology, University of PadovaPadovaItaly
| | - Ildiko Szabó
- Department of Biology, University of PadovaPadovaItaly
| | - Antonio Felipe
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| |
Collapse
|
13
|
Bachmann M, Li W, Edwards MJ, Ahmad SA, Patel S, Szabo I, Gulbins E. Voltage-Gated Potassium Channels as Regulators of Cell Death. Front Cell Dev Biol 2020; 8:611853. [PMID: 33381507 PMCID: PMC7767978 DOI: 10.3389/fcell.2020.611853] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Ion channels allow the flux of specific ions across biological membranes, thereby determining ion homeostasis within the cells. Voltage-gated potassium-selective ion channels crucially contribute to the setting of the plasma membrane potential, to volume regulation and to the physiologically relevant modulation of intracellular potassium concentration. In turn, these factors affect cell cycle progression, proliferation and apoptosis. The present review summarizes our current knowledge about the involvement of various voltage-gated channels of the Kv family in the above processes and discusses the possibility of their pharmacological targeting in the context of cancer with special emphasis on Kv1.1, Kv1.3, Kv1.5, Kv2.1, Kv10.1, and Kv11.1.
Collapse
Affiliation(s)
- Magdalena Bachmann
- Department of Biology, University of Padova, Padua, Italy.,Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Weiwei Li
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Michael J Edwards
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Syed A Ahmad
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Sameer Patel
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy.,Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padua, Italy
| | - Erich Gulbins
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
Bozic I, Savic D, Milosevic A, Janjic M, Laketa D, Tesovic K, Bjelobaba I, Jakovljevic M, Nedeljkovic N, Pekovic S, Lavrnja I. The Potassium Channel Kv1.5 Expression Alters During Experimental Autoimmune Encephalomyelitis. Neurochem Res 2019; 44:2733-2745. [PMID: 31624998 DOI: 10.1007/s11064-019-02892-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/23/2019] [Accepted: 10/11/2019] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory, neurodegenerative disease with an autoimmune component. It was suggested that potassium channels, which are involved in crucial biological functions may have a role in different diseases, including MS and its animal model, experimental autoimmune encephalomyelitis (EAE). It was shown that voltage-gated potassium channels Kv1.5 are responsible for fine-tuning in the immune physiology and influence proliferation and differentiation in microglia and astrocytes. Here, we explored the cellular distribution of the Kv1.5 channel, together with its transcript and protein expression in the male rat spinal cord during different stages of EAE. Our results reveal a decrease of Kv1.5 transcript and protein level at the peak of disease, where massive infiltration of myeloid cells occurs, together with reactive astrogliosis and demyelination. Also, we revealed that the presence of this channel is not found in infiltrating macrophages/microglia during EAE. It is interesting to note that Kv1.5 channel is expressed only in resting microglia in the naïve animals. Predominant expression of Kv1.5 channel was found in the astrocytes in all experimental groups, while some vimentin+ cells, resembling macrophages, are devoid of Kv1.5 expression. Our results point to the possible link between Kv1.5 channel and the pathophysiological processes in EAE.
Collapse
Affiliation(s)
- I Bozic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Savic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - A Milosevic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - M Janjic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Laketa
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - K Tesovic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - I Bjelobaba
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - M Jakovljevic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - N Nedeljkovic
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - S Pekovic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - I Lavrnja
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
15
|
Hernandez-Resendiz I, Hartung F, Pardo LA. Antibodies Targeting K V Potassium Channels: A Promising Treatment for Cancer. Bioelectricity 2019; 1:180-187. [PMID: 34471820 DOI: 10.1089/bioe.2019.0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated potassium channels are transmembrane proteins that allow flow of potassium across the membrane to regulate ion homeostasis, cell proliferation, migration, cell volume, and specific processes such as muscular contraction. Aberrant function or expression of potassium channels can underlie pathologies ranging from heart arrhythmia to cancer; the expression of potassium channels is altered in many types of cancer and that alteration correlates with malignancy and poor prognosis. Targeting potassium channels therefore constitutes a promising approach for cancer therapy. In this review, we discuss strategies to target a particular family of potassium channels, the voltage-gated potassium channels (KV) where a reasonable structural understanding is available. We also discuss the possible obstacles and advantages of such a strategy.
Collapse
Affiliation(s)
| | - Franziska Hartung
- AG Oncophysiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Luis A Pardo
- AG Oncophysiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
16
|
Implication of Voltage-Gated Potassium Channels in Neoplastic Cell Proliferation. Cancers (Basel) 2019; 11:cancers11030287. [PMID: 30823672 PMCID: PMC6468671 DOI: 10.3390/cancers11030287] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/21/2019] [Accepted: 02/24/2019] [Indexed: 12/12/2022] Open
Abstract
Voltage-gated potassium channels (Kv) are the largest group of ion channels. Kv are involved in controlling the resting potential and action potential duration in the heart and brain. Additionally, these proteins participate in cell cycle progression as well as in several other important features in mammalian cell physiology, such as activation, differentiation, apoptosis, and cell volume control. Therefore, Kv remarkably participate in the cell function by balancing responses. The implication of Kv in physiological and pathophysiological cell growth is the subject of study, as Kv are proposed as therapeutic targets for tumor regression. Though it is widely accepted that Kv channels control proliferation by allowing cell cycle progression, their role is controversial. Kv expression is altered in many cancers, and their participation, as well as their use as tumor markers, is worthy of effort. There is an ever-growing list of Kv that remodel during tumorigenesis. This review focuses on the actual knowledge of Kv channel expression and their relationship with neoplastic proliferation. In this work, we provide an update of what is currently known about these proteins, thereby paving the way for a more precise understanding of the participation of Kv during cancer development.
Collapse
|
17
|
Franklin BM, Voss SR, Osborn JL. Ion channel signaling influences cellular proliferation and phagocyte activity during axolotl tail regeneration. Mech Dev 2017; 146:42-54. [PMID: 28603004 PMCID: PMC6386162 DOI: 10.1016/j.mod.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 01/30/2023]
Abstract
Little is known about the potential for ion channels to regulate cellular behaviors during tissue regeneration. Here, we utilized an amphibian tail regeneration assay coupled with a chemical genetic screen to identify ion channel antagonists that altered critical cellular processes during regeneration. Inhibition of multiple ion channels either partially (anoctamin1/Tmem16a, anoctamin2/Tmem16b, KV2.1, KV2.2, L-type CaV channels and H/K ATPases) or completely (GlyR, GABAAR, KV1.5 and SERCA pumps) inhibited tail regeneration. Partial inhibition of tail regeneration by blocking the calcium activated chloride channels, anoctamin1&2, was associated with a reduction of cellular proliferation in tail muscle and mesenchymal regions. Inhibition of anoctamin 1/2 also altered the post-amputation transcriptional response of p44/42 MAPK signaling pathway genes, including decreased expression of erk1/erk2. We also found that complete inhibition via voltage gated K+ channel blockade was associated with diminished phagocyte recruitment to the amputation site. The identification of H+ pumps as required for axolotl tail regeneration supports findings in Xenopus and Planaria models, and more generally, the conservation of ion channels as regulators of tissue regeneration. This study provides a preliminary framework for an in-depth investigation of the mechanistic role of ion channels and their potential involvement in regulating cellular proliferation and other processes essential to wound healing, appendage regeneration, and tissue repair.
Collapse
Affiliation(s)
- Brandon M Franklin
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States
| | - S Randal Voss
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States
| | - Jeffrey L Osborn
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
18
|
de la Cruz A, Vera-Zambrano A, Peraza DA, Valenzuela C, Zapata JM, Perez-Chacon G, Gonzalez T. Fludarabine Inhibits K V1.3 Currents in Human B Lymphocytes. Front Pharmacol 2017; 8:177. [PMID: 28408885 PMCID: PMC5374215 DOI: 10.3389/fphar.2017.00177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/16/2017] [Indexed: 11/13/2022] Open
Abstract
Fludarabine (F-ara-A) is a purine analog commonly used in the treatment of indolent B cell malignancies that interferes with different aspects of DNA and RNA synthesis. KV1.3 K+ channels are membrane proteins involved in the maintenance of K+ homeostasis and the resting potential of the cell, thus controlling signaling events, proliferation and apoptosis in lymphocytes. Here we show that F-ara-A inhibits KV currents in human B lymphocytes. Our data indicate that KV1.3 is expressed in both BL2 and Dana B cell lines, although total KV1.3 levels were higher in BL2 than in Dana cells. However, KV currents in the plasma membrane were similar in both cell lines and were abrogated by the specific KV1.3 channel inhibitor PAP-1, indicating that KV1.3 accounts for most of the KV currents in these cell lines. F-ara-A, at a concentration (3.5 μM) similar to that achieved in the plasma of fludarabine phosphate-treated patients (3 μM), inhibited KV1.3 currents by 61 ± 6.3% and 52.3 ± 6.3% in BL2 and Dana B cells, respectively. The inhibitory effect of F-ara-A was concentration-dependent and showed an IC50 value of 0.36 ± 0.04 μM and a nH value of 1.07 ± 0.15 in BL2 cells and 0.34 ± 0.13 μM (IC50 ) and 0.77 ± 0.11 (nH ) in Dana cells. F-ara-A inhibition of plasma membrane KV1.3 was observed irrespective of its cytotoxic effect on the cells, BL2 cells being sensitive and Dana cells resistant to F-ara-A cytotoxicity. Interestingly, PAP-1, at concentrations as high as 10 μM, did not affect the viability of BL2 and Dana cells, indicating that blockage of KV1.3 in these cells is not toxic. Finally, F-ara-A had no effect on ectopically expressed KV1.3 channels, suggesting an indirect mechanism of current inhibition. In summary, our results describe the inhibitory effect of F-ara-A on the activity of KV1.3 channel. Although KV1.3 inhibition is not sufficient to induce cell death, further research is needed to determine whether it might still contribute to F-ara-A cytotoxicity in sensitive cells or be accountable for some of the clinical side effects of the drug.
Collapse
Affiliation(s)
- Alicia de la Cruz
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain
| | - Alba Vera-Zambrano
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain.,Departamento de Bioquímica, Universidad Autónoma de MadridMadrid, Spain
| | - Diego A Peraza
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain
| | - Juan M Zapata
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPaz)Madrid, Spain
| | - Gema Perez-Chacon
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPaz)Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de MadridMadrid, Spain.,Departamento de Bioquímica, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPaz)Madrid, Spain
| |
Collapse
|
19
|
Moral-Sanz J, Mahmoud AD, Ross FA, Eldstrom J, Fedida D, Hardie DG, Evans AM. AMP-activated protein kinase inhibits Kv 1.5 channel currents of pulmonary arterial myocytes in response to hypoxia and inhibition of mitochondrial oxidative phosphorylation. J Physiol 2016; 594:4901-15. [PMID: 27062501 PMCID: PMC5009768 DOI: 10.1113/jp272032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/26/2016] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS Progression of hypoxic pulmonary hypertension is thought to be due, in part, to suppression of voltage-gated potassium channels (Kv ) in pulmonary arterial smooth muscle by hypoxia, although the precise molecular mechanisms have been unclear. AMP-activated protein kinase (AMPK) has been proposed to couple inhibition of mitochondrial metabolism by hypoxia to acute hypoxic pulmonary vasoconstriction and progression of pulmonary hypertension. Inhibition of complex I of the mitochondrial electron transport chain activated AMPK and inhibited Kv 1.5 channels in pulmonary arterial myocytes. AMPK activation by 5-aminoimidazole-4-carboxamide riboside, A769662 or C13 attenuated Kv 1.5 currents in pulmonary arterial myocytes, and this effect was non-additive with respect to Kv 1.5 inhibition by hypoxia and mitochondrial poisons. Recombinant AMPK phosphorylated recombinant human Kv 1.5 channels in cell-free assays, and inhibited K(+) currents when introduced into HEK 293 cells stably expressing Kv 1.5. These results suggest that AMPK is the primary mediator of reductions in Kv 1.5 channels following inhibition of mitochondrial oxidative phosphorylation during hypoxia and by mitochondrial poisons. ABSTRACT Progression of hypoxic pulmonary hypertension is thought to be due, in part, to suppression of voltage-gated potassium channels (Kv ) in pulmonary arterial smooth muscle cells that is mediated by the inhibition of mitochondrial oxidative phosphorylation. We sought to determine the role in this process of the AMP-activated protein kinase (AMPK), which is intimately coupled to mitochondrial function due to its activation by LKB1-dependent phosphorylation in response to increases in the cellular AMP:ATP and/or ADP:ATP ratios. Inhibition of complex I of the mitochondrial electron transport chain using phenformin activated AMPK and inhibited Kv currents in pulmonary arterial myocytes, consistent with previously reported effects of mitochondrial inhibitors. Myocyte Kv currents were also markedly inhibited upon AMPK activation by A769662, 5-aminoimidazole-4-carboxamide riboside and C13 and by intracellular dialysis from a patch-pipette of activated (thiophosphorylated) recombinant AMPK heterotrimers (α2β2γ1 or α1β1γ1). Hypoxia and inhibitors of mitochondrial oxidative phosphorylation reduced AMPK-sensitive K(+) currents, which were also blocked by the selective Kv 1.5 channel inhibitor diphenyl phosphine oxide-1 but unaffected by the presence of the BKCa channel blocker paxilline. Moreover, recombinant human Kv 1.5 channels were phosphorylated by AMPK in cell-free assays, and K(+) currents carried by Kv 1.5 stably expressed in HEK 293 cells were inhibited by intracellular dialysis of AMPK heterotrimers and by A769662, the effects of which were blocked by compound C. We conclude that AMPK mediates Kv channel inhibition by hypoxia in pulmonary arterial myocytes, at least in part, through phosphorylation of Kv 1.5 and/or an associated protein.
Collapse
Affiliation(s)
- Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Fiona A Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - Jodene Eldstrom
- Department of Anaesthesiology. Pharmacology and Therapeutics, University of British Columbia, 2350 Health Science Mall, Vancouver, Canada, V6T 1Z3
| | - David Fedida
- Department of Anaesthesiology. Pharmacology and Therapeutics, University of British Columbia, 2350 Health Science Mall, Vancouver, Canada, V6T 1Z3
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| |
Collapse
|
20
|
Expression of KCNA5 Protein in Human Mammary Epithelial Cell Line Associated with Caveolin-1. J Membr Biol 2016; 249:449-57. [DOI: 10.1007/s00232-016-9885-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/06/2016] [Indexed: 11/24/2022]
|
21
|
Chimote AA, Hajdu P, Kottyan LC, Harley JB, Yun Y, Conforti L. Nanovesicle-targeted Kv1.3 knockdown in memory T cells suppresses CD40L expression and memory phenotype. J Autoimmun 2016; 69:86-93. [PMID: 26994905 DOI: 10.1016/j.jaut.2016.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 02/06/2023]
Abstract
Ca(2+) signaling controls activation and effector functions of T lymphocytes. Ca(2+) levels also regulate NFAT activation and CD40 ligand (CD40L) expression in T cells. CD40L in activated memory T cells binds to its cognate receptor, CD40, on other cell types resulting in the production of antibodies and pro-inflammatory mediators. The CD40L/CD40 interaction is implicated in the pathogenesis of autoimmune disorders and CD40L is widely recognized as a therapeutic target. Ca(2+) signaling in T cells is regulated by Kv1.3 channels. We have developed lipid nanoparticles that deliver Kv1.3 siRNAs (Kv1.3-NPs) selectively to CD45RO(+) memory T cells and reduce the activation-induced Ca(2+) influx. Herein we report that Kv1.3-NPs reduced NFAT activation and CD40L expression exclusively in CD45RO(+) T cells. Furthermore, Kv1.3-NPs suppressed cytokine release and induced a phenotype switch of T cells from predominantly memory to naïve. These findings indicate that Kv1.3-NPs operate as targeted immune suppressive agents with promising therapeutic potentials.
Collapse
Affiliation(s)
- Ameet A Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, USA
| | - Peter Hajdu
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John B Harley
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; US Department of Veterans Affairs Medical Center, Cincinnati, OH, USA
| | - Yeoheung Yun
- North Carolina A & T State University, Chemical, Biological and Bioengineering Department, Greensboro, NC, USA
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
22
|
Pérez-Verdaguer M, Capera J, Serrano-Novillo C, Estadella I, Sastre D, Felipe A. The voltage-gated potassium channel Kv1.3 is a promising multitherapeutic target against human pathologies. Expert Opin Ther Targets 2015; 20:577-91. [DOI: 10.1517/14728222.2016.1112792] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
23
|
Pfeifer M, Zheng B, Erdmann T, Koeppen H, McCord R, Grau M, Staiger A, Chai A, Sandmann T, Madle H, Dörken B, Chu YW, Chen AI, Lebovic D, Salles GA, Czuczman MS, Palanca-Wessels MC, Press OW, Advani R, Morschhauser F, Cheson BD, Lenz P, Ott G, Polson AG, Mundt KE, Lenz G. Anti-CD22 and anti-CD79B antibody drug conjugates are active in different molecular diffuse large B-cell lymphoma subtypes. Leukemia 2015; 29:1578-86. [PMID: 25708834 DOI: 10.1038/leu.2015.48] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 01/08/2015] [Accepted: 02/13/2015] [Indexed: 12/16/2022]
Abstract
Antibody drug conjugates (ADCs), in which cytotoxic drugs are linked to antibodies targeting antigens on tumor cells, represent promising novel agents for the treatment of malignant lymphomas. Pinatuzumab vedotin is an anti-CD22 ADC and polatuzumab vedotin an anti-CD79B ADC that are both linked to the microtubule-disrupting agent monomethyl auristatin E (MMAE). In the present study, we analyzed the activity of these agents in different molecular subtypes of diffuse large B-cell lymphoma (DLBCL) both in vitro and in early clinical trials. Both anti-CD22-MMAE and anti-CD79B-MMAE were highly active and induced cell death in the vast majority of activated B-cell-like (ABC) and germinal center B-cell-like (GCB) DLBCL cell lines. Similarly, both agents induced cytotoxicity in models with and without mutations in the signaling molecule CD79B. In line with these observations, relapsed and refractory DLBCL patients of both subtypes responded to these agents. Importantly, a strong correlation between CD22 and CD79B expression in vitro and in vivo was not detectable, indicating that patients should not be excluded from anti-CD22-MMAE or anti-CD79B-MMAE treatment because of low target expression. In summary, these studies suggest that pinatuzumab vedotin and polatuzumab vedotin are active agents for the treatment of patients with different subtypes of DLBCL.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- CD79 Antigens/genetics
- CD79 Antigens/immunology
- Cell Cycle/drug effects
- Cell Proliferation/drug effects
- Clinical Trials, Phase I as Topic
- Cohort Studies
- Flow Cytometry
- Follow-Up Studies
- Humans
- Immunoconjugates/pharmacology
- Immunoenzyme Techniques
- Lymphoma, Large B-Cell, Diffuse/classification
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mutation/genetics
- Neoplasm Staging
- Prognosis
- Sialic Acid Binding Ig-like Lectin 2/genetics
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M Pfeifer
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Germany
| | - B Zheng
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - T Erdmann
- 1] Division of Translational Oncology, Department of Medicine A, University Hospital Münster, Münster, Germany [2] Cluster of Excellence EXC 1003, Cells in Motion Münster, Germany
| | - H Koeppen
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - R McCord
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - M Grau
- Department of Physics, Philipps-University, Marburg, Germany
| | - A Staiger
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - A Chai
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - T Sandmann
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - H Madle
- 1] Division of Translational Oncology, Department of Medicine A, University Hospital Münster, Münster, Germany [2] Cluster of Excellence EXC 1003, Cells in Motion Münster, Germany
| | - B Dörken
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Germany
| | - Y-W Chu
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - A I Chen
- Department of Hematology-Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - D Lebovic
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - G A Salles
- Hematology Department, Hospices Civils de Lyon - Université de Lyon, Pierre-Bénite, France
| | - M S Czuczman
- Department of Medicine and Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - M C Palanca-Wessels
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Seattle Genetics Inc, Bothell, WA, USA
| | - O W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - R Advani
- Stanford University Medical Center, Stanford University, Stanford, CA, USA
| | - F Morschhauser
- Department of Hematology, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - B D Cheson
- Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington DC, USA
| | - P Lenz
- Department of Physics, Philipps-University, Marburg, Germany
| | - G Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - A G Polson
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - K E Mundt
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - G Lenz
- 1] Division of Translational Oncology, Department of Medicine A, University Hospital Münster, Münster, Germany [2] Cluster of Excellence EXC 1003, Cells in Motion Münster, Germany
| |
Collapse
|
24
|
Involvement of potassium channels in the progression of cancer to a more malignant phenotype. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2477-92. [PMID: 25517985 DOI: 10.1016/j.bbamem.2014.12.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 12/01/2014] [Accepted: 12/08/2014] [Indexed: 12/22/2022]
Abstract
Potassium channels are a diverse group of pore-forming transmembrane proteins that selectively facilitate potassium flow through an electrochemical gradient. They participate in the control of the membrane potential and cell excitability in addition to different cell functions such as cell volume regulation, proliferation, cell migration, angiogenesis as well as apoptosis. Because these physiological processes are essential for the correct cell function, K+ channels have been associated with a growing number of diseases including cancer. In fact, different K+ channel families such as the voltage-gated K+ channels, the ether à-go-go K+ channels, the two pore domain K+ channels and the Ca2+-activated K+ channels have been associated to tumor biology. Potassium channels have a role in neoplastic cell-cycle progression and their expression has been found abnormal in many types of tumors and cancer cells. In addition, the expression and activity of specific K+ channels have shown a significant correlation with the tumor malignancy grade. The aim of this overview is to summarize published data on K+ channels that exhibit oncogenic properties and have been linked to a more malignant cancer phenotype. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
|
25
|
Simma N, Bose T, Kahlfuss S, Mankiewicz J, Lowinus T, Lühder F, Schüler T, Schraven B, Heine M, Bommhardt U. NMDA-receptor antagonists block B-cell function but foster IL-10 production in BCR/CD40-activated B cells. Cell Commun Signal 2014; 12:75. [PMID: 25477292 PMCID: PMC4269920 DOI: 10.1186/s12964-014-0075-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 11/12/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND B cells are important effectors and regulators of adaptive and innate immune responses, inflammation and autoimmunity, for instance in anti-NMDA-receptor (NMDAR) encephalitis. Thus, pharmacological modulation of B-cell function could be an effective regimen in therapeutic strategies. Since the non-competitive NMDAR antagonist memantine is clinically applied to treat advanced Alzheimer`s disease and ketamine is supposed to improve the course of resistant depression, it is important to know how these drugs affect B-cell function. RESULTS Non-competitive NMDAR antagonists impaired B-cell receptor (BCR)- and lipopolysaccharide (LPS)-induced B-cell proliferation, reduced B-cell migration towards the chemokines SDF-1α and CCL21 and downregulated IgM and IgG secretion. Mechanistically, these effects were mediated through a blockade of Kv1.3 and KCa3.1 potassium channels and resulted in an attenuated Ca(2+)-flux and activation of Erk1/2, Akt and NFATc1. Interestingly, NMDAR antagonist treatment increased the frequency of IL-10 producing B cells after BCR/CD40 stimulation. CONCLUSIONS Non-competitive NMDAR antagonists attenuate BCR and Toll-like receptor 4 (TLR4) B-cell signaling and effector function and can foster IL-10 production. Consequently, NMDAR antagonists may be useful to target B cells in autoimmune diseases or pathological systemic inflammation. The drugs' additional side effects on B cells should be considered in treatments of neuronal disorders with NMDAR antagonists.
Collapse
Affiliation(s)
- Narasimhulu Simma
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Tanima Bose
- RG Molecular Physiology, Leibniz Institute of Neurobiology, Brenneckestr. 6, 39118, Magdeburg, Germany.
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Judith Mankiewicz
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Fred Lühder
- Department of Neuroimmunology, Institute for Multiple Sclerosis Research and The Hertie Foundation, Waldweg 33, 37073, Göttingen, Germany.
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Department of Immune Control, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany.
| | - Martin Heine
- RG Molecular Physiology, Leibniz Institute of Neurobiology, Brenneckestr. 6, 39118, Magdeburg, Germany.
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| |
Collapse
|
26
|
Ryland KE, Svoboda LK, Vesely ED, McIntyre JC, Zhang L, Martens JR, Lawlor ER. Polycomb-dependent repression of the potassium channel-encoding gene KCNA5 promotes cancer cell survival under conditions of stress. Oncogene 2014; 34:4591-600. [PMID: 25435365 PMCID: PMC4451446 DOI: 10.1038/onc.2014.384] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/23/2014] [Accepted: 10/10/2014] [Indexed: 02/08/2023]
Abstract
Relapse after clinical remission remains a leading cause of cancer-associated death. Although the mechanisms of tumor relapse are complex, the ability of cancer cells to survive physiological stress is a prerequisite for recurrence. Ewing sarcoma (ES) and neuroblastoma (NB) are aggressive cancers that frequently relapse after initial remission. In addition, both tumors overexpress the polycomb group (PcG) proteins BMI-1 and EZH2, which contribute to tumorigenicity. We have discovered that ES and NB resist hypoxic stress-induced death and that survival depends on PcG function. Epigenetic repression of developmental programs is the most well-established cancer-associated function of PcG proteins. However, we noted that voltage-gated potassium (Kv) channel genes are also targets of PcG regulation in stem cells. Given the role of potassium in regulating apoptosis, we reasoned that repression of Kv channel genes might have a role in cancer cell survival. Here we describe our novel finding that PcG-dependent repression of the Kv1.5 channel gene KCNA5 contributes to cancer cell survival under conditions of stress. We show that survival of cancer cells in stress is dependent upon suppression of Kv1.5 channel function. The KCNA5 promoter is marked in cancer cells with PcG-dependent chromatin repressive modifications that increase in hypoxia. Genetic and pharmacological inhibition of BMI-1 and EZH2, respectively, restore KCNA5 expression, which sensitizes cells to stress-induced death. In addition, ectopic expression of the Kv1.5 channel induces apoptotic cell death under conditions of hypoxia. These findings identify a novel role for PcG proteins in promoting cancer cell survival via repression of KCNA5.
Collapse
Affiliation(s)
- K E Ryland
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.,Translational Oncology Program, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - L K Svoboda
- Translational Oncology Program, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - E D Vesely
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - J C McIntyre
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - L Zhang
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - J R Martens
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - E R Lawlor
- Translational Oncology Program, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
27
|
Lee BH, Ryu PD, Lee SY. Serum starvation-induced voltage-gated potassium channel Kv7.5 expression and its regulation by Sp1 in canine osteosarcoma cells. Int J Mol Sci 2014; 15:977-93. [PMID: 24434641 PMCID: PMC3907850 DOI: 10.3390/ijms15010977] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/20/2013] [Accepted: 12/30/2013] [Indexed: 01/03/2023] Open
Abstract
The KCNQ gene family, whose members encode Kv7 channels, belongs to the voltage-gated potassium (Kv) channel group. The roles of this gene family have been widely investigated in nerve and muscle cells. In the present study, we investigated several characteristics of Kv7.5, which is strongly expressed in the canine osteosarcoma cell line, CCL-183. Serum starvation upregulated Kv7.5 expression, and the Kv7 channel opener, flupirtine, attenuated cell proliferation by arresting cells in the G0/G1 phase. We also showed that Kv7.5 knockdown helps CCL-183 cells to proliferate. In an effort to find an endogenous regulator of Kv7.5, we used mithramycin A to reduce the level of the transcription factor Sp1, and it strongly inhibited the induction of Kv7.5 in CCL-183 cells. These results suggest that the activation of Kv7.5 by flupirtine may exert an anti-proliferative effect in canine osteosarcoma. Therefore, Kv7.5 is a possible molecular target for canine osteosarcoma therapy.
Collapse
Affiliation(s)
- Bo Hyung Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - Pan Dong Ryu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| |
Collapse
|
28
|
Comes N, Bielanska J, Vallejo-Gracia A, Serrano-Albarrás A, Marruecos L, Gómez D, Soler C, Condom E, Ramón Y Cajal S, Hernández-Losa J, Ferreres JC, Felipe A. The voltage-dependent K(+) channels Kv1.3 and Kv1.5 in human cancer. Front Physiol 2013; 4:283. [PMID: 24133455 PMCID: PMC3794381 DOI: 10.3389/fphys.2013.00283] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
Voltage-dependent K+ channels (Kv) are involved in a number of physiological processes, including immunomodulation, cell volume regulation, apoptosis as well as differentiation. Some Kv channels participate in the proliferation and migration of normal and tumor cells, contributing to metastasis. Altered expression of Kv1.3 and Kv1.5 channels has been found in several types of tumors and cancer cells. In general, while the expression of Kv1.3 apparently exhibits no clear pattern, Kv1.5 is induced in many of the analyzed metastatic tissues. Interestingly, evidence indicates that Kv1.5 channel shows inversed correlation with malignancy in some gliomas and non-Hodgkin's lymphomas. However, Kv1.3 and Kv1.5 are similarly remodeled in some cancers. For instance, expression of Kv1.3 and Kv1.5 correlates with a certain grade of tumorigenicity in muscle sarcomas. Differential remodeling of Kv1.3 and Kv1.5 expression in human cancers may indicate their role in tumor growth and their importance as potential tumor markers. However, despite of this increasing body of information, which considers Kv1.3 and Kv1.5 as emerging tumoral markers, further research must be performed to reach any conclusion. In this review, we summarize what it has been lately documented about Kv1.3 and Kv1.5 channels in human cancer.
Collapse
Affiliation(s)
- Núria Comes
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|