1
|
Kim CW, Kim EJ, Woo MS, Cao DL, Cirunduzi AC, Ryu JH, Kong IK, Lee DK, Hong SG, Han J, Kang D. Downregulation of TASK-3 Channel Induces Senescence in Granulosa Cells of Bovine Cystic Ovarian Follicles. Int J Mol Sci 2024; 25:10199. [PMID: 39337686 PMCID: PMC11432027 DOI: 10.3390/ijms251810199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian cysts are linked to hormone imbalances and altered gene expressions, but the connection between cysts and ion channel expression is understudied. This study explored the role of TWIK-related acid-sensitive K+ (TASK) channels in bovine ovarian cyst formation. The ovarian follicles were split into small (5 to 10 mm in diameter) and large (>25 mm in diameter) groups. Among the measured K+, Na+, and Cl- concentrations in follicular fluid (FF) obtained from small-sized follicles (SFs) and large-sized follicles (LFs), the K+ concentration was significantly lower in LFFF. Quantitative PCR, Western blot, and immunocytochemistry data revealed that TASK-3 expression levels significantly decreased by approximately 50% in LFs and granulosa cells obtained from LFs (LFGCs) compared to the corresponding controls. The TASK-3 protein was localized to the plasma membranes of GCs. The diameters of LFGCs were larger than those of SFGCs. The cell swelling response to exposure to a hypotonic solution (200 mOsm/L) was highly reduced in TASK-3-overexpressing cells compared to vector-transfected cells. TASK-3-knockdown cells showed arrested growth. Senescence markers were detected in LFGCs and TASK-3-knockdown cells. These findings suggest that reduced TASK-3 expression in LFs is associated with the inhibition of GC growth, leading to senescence and cyst formation.
Collapse
Affiliation(s)
- Chang-Woon Kim
- Department of Obstetrics and Gynecology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Eun-Jin Kim
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Min Seok Woo
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Dang Long Cao
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Asifiwe Clarisse Cirunduzi
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Ji Hyeon Ryu
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Il-Keun Kong
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Dong Kun Lee
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Seong-Geun Hong
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jaehee Han
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Dawon Kang
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| |
Collapse
|
2
|
Rinné S, Schick F, Vowinkel K, Schütte S, Krasel C, Kauferstein S, Schäfer MKH, Kiper AK, Müller T, Decher N. Potassium channel TASK-5 forms functional heterodimers with TASK-1 and TASK-3 to break its silence. Nat Commun 2024; 15:7548. [PMID: 39215006 PMCID: PMC11364637 DOI: 10.1038/s41467-024-51288-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
TASK-5 (KCNK15) belongs to the acid-sensitive subfamily of two-pore domain potassium (K2P) channels, which includes TASK-1 and TASK-3. TASK-5 stands out as K2P channel for which there is no functional data available, since it was reported in 2001 as non-functional and thus "silent". Here we show that TASK-5 channels are indeed non-functional as homodimers, but are involved in the formation of functional channel complexes with TASK-1 and TASK-3. TASK-5 negatively modulates the surface expression of TASK channels, while the heteromeric TASK-5-containing channel complexes located at the plasma membrane are characterized by changes in single-channel conductance, Gq-coupled receptor-mediated channel inhibition, and sensitivity to TASK modulators. The unique pharmacology of TASK-1/TASK-5 heterodimers, affected by a common polymorphism in KCNK15, needs to be carefully considered in the future development of drugs targeting TASK channels. Our observations provide an access to study TASK-5 at the functional level, particularly in malignant cancers associated with KCNK15.
Collapse
Affiliation(s)
- Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Florian Schick
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Kirsty Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Sven Schütte
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Cornelius Krasel
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Silke Kauferstein
- Centre for Sudden Cardiac Death and Institute of Legal Medicine, University Hospital Frankfurt, Goethe-University, Frankfurt/Main, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany
| | - Martin K-H Schäfer
- Institute of Anatomy and Cell Biology, Philipps University Marburg, Marburg, Germany
| | - Aytug K Kiper
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Thomas Müller
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
3
|
Hu Z, Jia Q, Yao S, Chen X. The TWIK-related acid sensitive potassium 3 (TASK-3) channel contributes to the different effects of anesthetics on the growth and metastasis of ovarian cancer cells. Heliyon 2024; 10:e34973. [PMID: 39161826 PMCID: PMC11332837 DOI: 10.1016/j.heliyon.2024.e34973] [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] [Received: 02/08/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/21/2024] Open
Abstract
Different anesthetics exert different effects on the long-term outcomes of various cancers. The TWIK-related acid sensitive potassium 3 (TASK-3) channel is an important target of anesthetics and is upregulated in various cancers. However, the role and underlying mechanism of TASK-3 channel in the effects of anesthetics on ovarian cancer remain unknown. Here, we tested whether the TASK-3 channel contributes to the effects of anesthetics on ovarian cancers. We found that the TASK-3 channel was overexpressed in human ovarian cancer and ovarian cancer cell lines. Clinically relevant concentrations of lidocaine, as a TASK-3 channel inhibitor, exert inhibitory effects on tumor growth and metastasis of ovarian cancer cells in vitro and in vivo, whereas the TASK-3 channel potent activator sevoflurane had protumor effects and propofol had no significant effects on tumor growth and metastasis of ovarian cancer. Knockdown of the TASK-3 channel by TASK-3 shRNA attenuated the effects of lidocaine and sevoflurane. Moreover, mitochondrial TASK-3 channel contributes to the effects of lidocaine and sevoflurane on the mitochondrial functions of ovarian cancer. Taken together, the TASK-3 channel, especially the mitochondrial TASK-3 (MitoTASK-3) channel, is a molecular substrate for the effects of lidocaine and sevoflurane on the tumor growth and metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Zhiqiang Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Qi Jia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| |
Collapse
|
4
|
Fan X, Ye Y, Saha A, Peng L, Pindi C, Wang Q, Yang L, Liu J, Tang X, Palermo G, Liao J, Xu T, Lu Y, Du G. Fine-tuning pH sensor H98 by remote essential residues in the hydrogen-bond network of mTASK-3. Int J Biol Macromol 2024; 273:132892. [PMID: 38878921 PMCID: PMC11583903 DOI: 10.1016/j.ijbiomac.2024.132892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/21/2024] [Accepted: 06/02/2024] [Indexed: 06/23/2024]
Abstract
TASK-3 generates a background K+ conductance which when inhibited by acidification depolarizes membrane potential and increases cell excitability. These channels sense pH by protonation of histidine residue H98, but recent evidence revealed that several other amino acid residues also contribute to TASK-3 pH sensitivity, suggesting that the pH sensitivity is determined by an intermolecular network. Here we use electrophysiology and molecular modeling to characterize the nature and requisite role(s) of multiple amino acids in pH sensing by TASK-3. Our results suggest that the pH sensor H98 and consequently pH sensitivity is influenced by remote amino acids that function as a hydrogen-bonding network to modulate ionic conductivity. Among the residues in the network, E30 and K79 are the most important for passing external signals near residue S31 to H98. The hydrogen-bond network plays a key role in selectivity or pH sensing in mTASK-3, and E30 and S31 in the network can modulate the conductive properties (E30) or reverse the pH sensitivity and selectivity of the channel (S31). Molecular dynamics simulations and pK1/2 calculation revealed that double mutants involving H98 + S31 primarily regulate the structure stability of the pore selectivity filter and pore loop regions, further strengthen the stability of the cradle suspension system, and alter the ionization state of E30 and K79, thereby preventing pore conformational change that normally occurs in response to varying extracellular pH. These results demonstrate that crucial residues in the hydrogen-bond network can remotely tune the pH sensing of mTASK-3 and may be a potential allosteric regulatory site for therapeutic molecule development.
Collapse
Affiliation(s)
- Xueming Fan
- Department of Pain Management, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Yifei Ye
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Aakash Saha
- Department of Bioengineering, University of California Riverside, 900 University Avenue, Riverside, CA 92521, United States
| | - Li Peng
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Chinmai Pindi
- Department of Bioengineering, University of California Riverside, 900 University Avenue, Riverside, CA 92521, United States
| | - Qi Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangdong Tang
- Sleep Medicine Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Giulia Palermo
- Department of Bioengineering, University of California Riverside, 900 University Avenue, Riverside, CA 92521, United States
| | - Jiayu Liao
- Department of Bioengineering, University of California Riverside, 900 University Avenue, Riverside, CA 92521, United States; Huaxi-Cal Research Center for Predictive Intervention Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Tingting Xu
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guizhou, Guangdong 510530, China
| | - Yongzhi Lu
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, China.
| | - Guizhi Du
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
5
|
Xie G, Si Q, Zhang G, Fan Y, Li Q, Leng P, Qiao F, Liang S, Yu R, Wang Y. The role of imprinting genes' loss of imprints in cancers and their clinical implications. Front Oncol 2024; 14:1365474. [PMID: 38812777 PMCID: PMC11133587 DOI: 10.3389/fonc.2024.1365474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024] Open
Abstract
Genomic imprinting plays an important role in the growth and development of mammals. When the original imprint status of these genes is lost, known as loss of imprinting (LOI), it may affect growth, neurocognitive development, metabolism, and even tumor susceptibility. The LOI of imprint genes has gradually been found not only as an early event in tumorigenesis, but also to be involved in progression. More than 120 imprinted genes had been identified in humans. In this review, we summarized the most studied LOI of two gene clusters and 13 single genes in cancers. We focused on the roles they played, that is, as growth suppressors and anti-apoptosis agents, sustaining proliferative signaling or inducing angiogenesis; the molecular pathways they regulated; and especially their clinical significance. It is notable that 12 combined forms of multi-genes' LOI, 3 of which have already been used as diagnostic models, achieved good sensitivity, specificity, and accuracy. In addition, the methods used for LOI detection in existing research are classified into detection of biallelic expression (BAE), differentially methylated regions (DMRs), methylation, and single-nucleotide polymorphisms (SNPs). These all indicated that the detection of imprinting genes' LOI has potential clinical significance in cancer diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Guojing Xie
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin Si
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangjie Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Laboratory, Chengdu Fifth People’s Hospital, Chengdu, China
| | - Yu Fan
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, Chengdu, China
| | - Qinghua Li
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Leng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, Chengdu, China
| | - Fengling Qiao
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, Chengdu, China
| | - Simin Liang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Yu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, Chengdu, China
| | - Yingshuang Wang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, Chengdu, China
| |
Collapse
|
6
|
Chatelain FC, Gilbert N, Bichet D, Jauch A, Feliciangeli S, Lesage F, Bignucolo O. Mechanistic basis of the dynamic response of TWIK1 ionic selectivity to pH. Nat Commun 2024; 15:3849. [PMID: 38719838 PMCID: PMC11079055 DOI: 10.1038/s41467-024-48067-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Highly selective for K+ at neutral pH, the TWIK1 channel becomes permeable to Na+ upon acidification. Using molecular dynamics simulations, we identify a network of residues involved in this unique property. Between the open and closed states previously observed by electron microscopy, molecular dynamics simulations show that the channel undergoes conformational changes between pH 7.5-6 involving residues His122, Glu235, Lys246 and Phe109. A complex network of interactions surrounding the selectivity filter at high pH transforms into a simple set of stronger interactions at low pH. In particular, His122 protonated by acidification moves away from Lys246 and engages in a salt bridge with Glu235. In addition, stacking interactions between Phe109 and His122, which stabilize the selectivity filter in its K+-selective state at high pH, disappear upon acidification. This leads to dissociation of the Phe109 aromatic side chain from this network, resulting in the Na+-permeable conformation of the channel.
Collapse
Affiliation(s)
- Franck C Chatelain
- Université Côte d'Azur, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Institut de pharmacologie moléculaire et cellulaire, 06560, Valbonne, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
| | - Nicolas Gilbert
- Université Côte d'Azur, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Institut de pharmacologie moléculaire et cellulaire, 06560, Valbonne, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
| | - Delphine Bichet
- Université Côte d'Azur, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Institut de pharmacologie moléculaire et cellulaire, 06560, Valbonne, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
| | - Annaïse Jauch
- Immunodeficiency Laboratory, Department of Biomedicine, Basel, Switzerland
| | - Sylvain Feliciangeli
- Université Côte d'Azur, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Institut de pharmacologie moléculaire et cellulaire, 06560, Valbonne, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
| | - Florian Lesage
- Université Côte d'Azur, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Institut de pharmacologie moléculaire et cellulaire, 06560, Valbonne, France.
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France.
| | - Olivier Bignucolo
- Université Côte d'Azur, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Institut de pharmacologie moléculaire et cellulaire, 06560, Valbonne, France
- Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
7
|
Sakellakis M, Chalkias A. The Role οf Ion Channels in the Development and Progression of Prostate Cancer. Mol Diagn Ther 2023; 27:227-242. [PMID: 36600143 DOI: 10.1007/s40291-022-00636-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 01/06/2023]
Abstract
Ion channels have major regulatory functions in living cells. Apart from their role in ion transport, they are responsible for cellular electrogenesis and excitability, and may also regulate tissue homeostasis. Although cancer is not officially classified as a channelopathy, it has been increasingly recognized that ion channel aberrations play an important role in virtually all cancer types. Ion channels can exert pro-tumorigenic activities due to genetic or epigenetic alterations, or as a response to molecular signals, such as growth factors, hormones, etc. Increasing evidence suggests that ion channels and pumps play a critical role in the regulation of prostate cancer cell proliferation, apoptosis evasion, migration, epithelial-to-mesenchymal transition, and angiogenesis. There is also evidence suggesting that ion channels might play a role in treatment failure in patients with prostate cancer. Hence, they represent promising targets for diagnosis, staging, and treatment, and their effects may be of particular significance for specific patient populations, including those undergoing anesthesia and surgery. In this article, the role of major types of ion channels involved in the development and progression of prostate cancer are reviewed. Identifying the underlying molecular mechanisms of the pro-tumorigenic effects of ion channels may potentially inform the development of novel therapeutic strategies to counter this malignancy.
Collapse
Affiliation(s)
- Minas Sakellakis
- Hellenic GU Cancer Group, Athens, Greece. .,Department of Medical Oncology, Metropolitan Hospital, 9 Ethnarchou Makariou, 18547, Athens, Greece.
| | - Athanasios Chalkias
- Department of Anesthesiology, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Outcomes Research Consortium, Cleveland, OH, USA
| |
Collapse
|
8
|
Cheng Y, Tang Y, Tan Y, Li J, Zhang X. KCNK9 mediates the inhibitory effects of genistein on hepatic metastasis from colon cancer. Clinics (Sao Paulo) 2023; 78:100141. [PMID: 36905879 PMCID: PMC10019991 DOI: 10.1016/j.clinsp.2022.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/08/2022] [Accepted: 11/01/2022] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE The tyrosine-protein kinase inhibitor, genistein, can inhibit cell malignant transformation and has an antitumor effect on various types of cancer. It has been shown that both genistein and KNCK9 can inhibit colon cancer. This research aimed to investigate the suppressive effects of genistein on colon cancer cells and the association between the application of genistein and KCNK9 expression level. METHODS The Cancer Genome Atlas (TCGA) database was used to study the correlation between the KCNK9 expression level and the prognosis of colon cancer patients. HT29 and SW480 colon cancer cell lines were cultured to examine the inhibitory effects of KCNK9 and genistein on colon cancer in vitro, and a mouse model of colon cancer with liver metastasis was established to verify the inhibitory effect of genistein in vivo. RESULTS KCNK9 was overexpressed in colon cancer cells and was associated with a shorter Overall Survival (OS), a shorter Disease-Specific Survival (DFS), and a shorter Progression-Free Interval (PFI) of colon cancer patients. In vitro experiments showed that downregulation of KCNK9 or genistein application could suppress cell proliferation, migration, and invasion abilities, induce cell cycle quiescence, promote cell apoptosis, and reduce epithelial-mesenchymal transition of the colon cancer cell line. In vivo experiments revealed that silencing of KCNK9 or application of genistein could inhibit hepatic metastasis from colon cancer. Additionally, genistein could inhibit KCNK9 expression, thereby attenuating Wnt/β-catenin signaling pathway. CONCLUSION Genistein inhibited the occurrence and progression of colon cancer through Wnt/β-catenin signaling pathway that could be mediated by KCNK9.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Tang
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiming Tan
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Li
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuping Zhang
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, China.
| |
Collapse
|
9
|
Fan X, Lu Y, Du G, Liu J. Advances in the Understanding of Two-Pore Domain TASK Potassium Channels and Their Potential as Therapeutic Targets. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238296. [PMID: 36500386 PMCID: PMC9736439 DOI: 10.3390/molecules27238296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
TWIK-related acid-sensitive K+ (TASK) channels, including TASK-1, TASK-3, and TASK-5, are important members of the two-pore domain potassium (K2P) channel family. TASK-5 is not functionally expressed in the recombinant system. TASK channels are very sensitive to changes in extracellular pH and are active during all membrane potential periods. They are similar to other K2P channels in that they can create and use background-leaked potassium currents to stabilize resting membrane conductance and repolarize the action potential of excitable cells. TASK channels are expressed in both the nervous system and peripheral tissues, including excitable and non-excitable cells, and are widely engaged in pathophysiological phenomena, such as respiratory stimulation, pulmonary hypertension, arrhythmia, aldosterone secretion, cancers, anesthesia, neurological disorders, glucose homeostasis, and visual sensitivity. Therefore, they are important targets for innovative drug development. In this review, we emphasized the recent advances in our understanding of the biophysical properties, gating profiles, and biological roles of TASK channels. Given the different localization ranges and biologically relevant functions of TASK-1 and TASK-3 channels, the development of compounds that selectively target TASK-1 and TASK-3 channels is also summarized based on data reported in the literature.
Collapse
Affiliation(s)
- Xueming Fan
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Anesthesiology, Guizhou Provincial People’s Hospital, Guiyang 550002, China
| | - Yongzhi Lu
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510700, China
| | - Guizhi Du
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (G.D.); (J.L.)
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (G.D.); (J.L.)
| |
Collapse
|
10
|
Arévalo B, Bedoya M, Kiper AK, Vergara F, Ramírez D, Mazola Y, Bustos D, Zúñiga R, Cikutovic R, Cayo A, Rinné S, Ramirez-Apan MT, Sepúlveda FV, Cerda O, López-Collazo E, Decher N, Zúñiga L, Gutierrez M, González W. Selective TASK-1 Inhibitor with a Defined Structure–Activity Relationship Reduces Cancer Cell Proliferation and Viability. J Med Chem 2022; 65:15014-15027. [DOI: 10.1021/acs.jmedchem.1c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Bárbara Arévalo
- Centro de Estudios en Alimentos Procesados−CEAP, Conicyt, Programa Regional R19A10001, Gore Maule, 3460000 Talca, Chile
| | - Mauricio Bedoya
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, 3460000 Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, 3480094 Talca, Chile
| | - Aytug K. Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 1-2, 35037 Marburg, Germany
| | - Fernando Vergara
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, 4030000 Concepción, Chile
| | - Yuliet Mazola
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, 3460000 Talca, Chile
- Laboratorio de Bioinformática y Química Computacional (LBQC), Escuela de Química y Farmacia, Facultad de Medicina, Universidad Católica del Maule, 3460000 Talca, Chile
| | - Rafael Zúñiga
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
- Instituto de Investigación Interdisciplinaria, Vicerrectoría Académica, Universidad de Talca, 3460000 Talca, Chile
| | - Rocio Cikutovic
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
| | - Angel Cayo
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 1-2, 35037 Marburg, Germany
| | - M. Teresa Ramirez-Apan
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Coyoacán, 04510 México, DF, México
| | - Francisco V. Sepúlveda
- Centro de Estudios Científicos (CECs), 5110466 Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, 5110466 Valdivia, Chile
| | - Oscar Cerda
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, 8380453 Santiago, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Facultad de Medicina, Universidad de Chile, 8380453 Santiago, Chile
| | - Eduardo López-Collazo
- The Innate Immune Response Group and Tumor Immunology Laboratory, IdiPAZ, La Paz University Hospital, 8046 Madrid, Spain
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 1-2, 35037 Marburg, Germany
- Marburg Center for Mind, Brain and Behavior−MCMBB, Philipps-University Marburg, 35037 Marburg, Germany
| | - Leandro Zúñiga
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
| | - Margarita Gutierrez
- Laboratorio de Síntesis y Actividad Biológica, Instituto de Química de Recursos Naturales, Universidad de Talca, 1 poniente No. 1141, 3460000 Talca, Chile
| | - Wendy González
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
| |
Collapse
|
11
|
Zhao J, Li M, Xu J, Cheng W. The modulation of ion channels in cancer chemo-resistance. Front Oncol 2022; 12:945896. [PMID: 36033489 PMCID: PMC9399684 DOI: 10.3389/fonc.2022.945896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Ion channels modulate the flow of ions into and out of a cell or intracellular organelle, leading to generation of electrical or chemical signals and regulating ion homeostasis. The abundance of ion channels in the plasma and intracellular membranes are subject to physiological and pathological regulations. Abnormal and dysregulated expressions of many ion channels are found to be linked to cancer and cancer chemo-resistance. Here, we will summarize ion channels distribution in multiple tumors. And the involvement of ion channels in cancer chemo-resistance will be highlighted.
Collapse
|
12
|
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: 29] [Impact Index Per Article: 9.7] [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
|
13
|
Skaar DA, Dietze EC, Alva-Ornelas JA, Ann D, Schones DE, Hyslop T, Sistrunk C, Zalles C, Ambrose A, Kennedy K, Idassi O, Miranda Carboni G, Gould MN, Jirtle RL, Seewaldt VL. Epigenetic Dysregulation of KCNK9 Imprinting and Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:6031. [PMID: 34885139 PMCID: PMC8656495 DOI: 10.3390/cancers13236031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/26/2021] [Indexed: 12/02/2022] Open
Abstract
Genomic imprinting is an inherited form of parent-of-origin specific epigenetic gene regulation that is dysregulated by poor prenatal nutrition and environmental toxins. KCNK9 encodes for TASK3, a pH-regulated potassium channel membrane protein that is overexpressed in 40% of breast cancer. However, KCNK9 gene amplification accounts for increased expression in <10% of these breast cancers. Here, we showed that KCNK9 is imprinted in breast tissue and identified a differentially methylated region (DMR) controlling its imprint status. Hypomethylation at the DMR, coupled with biallelic expression of KCNK9, occurred in 63% of triple-negative breast cancers (TNBC). The association between hypomethylation and TNBC status was highly significant in African-Americans (p = 0.006), but not in Caucasians (p = 0.70). KCNK9 hypomethylation was also found in non-cancerous tissue from 77% of women at high-risk of developing breast cancer. Functional studies demonstrated that the KCNK9 gene product, TASK3, regulates mitochondrial membrane potential and apoptosis-sensitivity. In TNBC cells and non-cancerous mammary epithelial cells from high-risk women, hypomethylation of the KCNK9 DMR predicts for increased TASK3 expression and mitochondrial membrane potential (p < 0.001). This is the first identification of the KCNK9 DMR in mammary epithelial cells and demonstration that its hypomethylation in breast cancer is associated with increases in both mitochondrial membrane potential and apoptosis resistance. The high frequency of hypomethylation of the KCNK9 DMR in TNBC and non-cancerous breast tissue from high-risk women provides evidence that hypomethylation of the KNCK9 DMR/TASK3 overexpression may serve as a marker of risk and a target for prevention of TNBC, particularly in African American women.
Collapse
Affiliation(s)
- David A. Skaar
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA;
| | - Eric C. Dietze
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Jackelyn A. Alva-Ornelas
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - David Ann
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Dustin E. Schones
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Terry Hyslop
- Department of Biostatistics, School of Medicine, Duke University, Durham, NC 27710, USA;
| | - Christopher Sistrunk
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Carola Zalles
- Department of Pathology, Mercy Hospital, Miami, FL 33133, USA;
| | - Adrian Ambrose
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Kendall Kennedy
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Ombeni Idassi
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Gustavo Miranda Carboni
- Laboratory of Oncology, Department of Oncology, School of Medicine, University of Tennessee Health Science, Memphis, TN 38163, USA;
| | - Michael N. Gould
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Randy L. Jirtle
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA;
| | - Victoria L. Seewaldt
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| |
Collapse
|
14
|
Mathie A, Veale EL, Golluscio A, Holden RG, Walsh Y. Pharmacological Approaches to Studying Potassium Channels. Handb Exp Pharmacol 2021; 267:83-111. [PMID: 34195873 DOI: 10.1007/164_2021_502] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this review, we consider the pharmacology of potassium channels from the perspective of these channels as therapeutic targets. Firstly, we describe the three main families of potassium channels in humans and disease states where they are implicated. Secondly, we describe the existing therapeutic agents which act on potassium channels and outline why these channels represent an under-exploited therapeutic target with potential for future drug development. Thirdly, we consider the evidence desired in order to embark on a drug discovery programme targeting a particular potassium channel. We have chosen two "case studies": activators of the two-pore domain potassium (K2P) channel TREK-2 (K2P10.1), for the treatment of pain and inhibitors of the voltage-gated potassium channel KV1.3, for use in autoimmune diseases such as multiple sclerosis. We describe the evidence base to suggest why these are viable therapeutic targets. Finally, we detail the main technical approaches available to characterise the pharmacology of potassium channels and identify novel regulatory compounds. We draw particular attention to the Comprehensive in vitro Proarrhythmia Assay initiative (CiPA, https://cipaproject.org ) project for cardiac safety, as an example of what might be both desirable and possible in the future, for ion channel regulator discovery projects.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, University of Kent, Kent, UK. .,Medway School of Pharmacy, University of Greenwich, London, UK. .,School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, UK.
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Alessia Golluscio
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Robyn G Holden
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Yvonne Walsh
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| |
Collapse
|
15
|
Abstract
Monoclonal antibodies combine specificity and high affinity binding with excellent pharmacokinetic properties and are rapidly being developed for a wide range of drug targets including clinically important potassium ion channels. Nonetheless, while therapeutic antibodies come with great promise, K+ channels represent particularly difficult targets for biologics development for a variety of reasons that include their dynamic structures and relatively small extracellular loops, their high degree of sequence conservation (leading to immune tolerance), and their generally low-level expression in vivo. The process is made all the more difficult when large numbers of antibody candidates must be screened for a given target, or when lead candidates fail to cross-react with orthologous channels in animal disease models due to their highly selective binding properties. While the number of antibodies targeting potassium channels in preclinical or clinical development is still modest, significant advances in the areas of protein expression and antibody screening are converging to open the field to an avalanche of new drugs. Here, the opportunities and constraints associated with the discovery of antibodies against K+ channels are discussed, with an emphasis on novel technologies that are opening the field to exciting new possibilities for biologics development.
Collapse
|
16
|
Levin M. Bioelectrical approaches to cancer as a problem of the scaling of the cellular self. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 165:102-113. [PMID: 33961843 DOI: 10.1016/j.pbiomolbio.2021.04.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
One lens with which to understand the complex phenomenon of cancer is that of developmental biology. Cancer is the inevitable consequence of a breakdown of the communication that enables individual cells to join into computational networks that work towards large-scale, morphogenetic goals instead of more primitive, unicellular objectives. This perspective suggests that cancer may be a physiological disorder, not necessarily due to problems with the genetically-specified protein hardware. One aspect of morphogenetic coordination is bioelectric signaling, and indeed an abnormal bioelectric signature non-invasively reveals the site of incipient tumors in amphibian models. Functionally, a disruption of resting potential states triggers metastatic melanoma phenotypes in embryos with no genetic defects or carcinogen exposure. Conversely, optogenetic or molecular-biological modulation of bioelectric states can override powerful oncogenic mutations and prevent or normalize tumors. The bioelectrically-mediated information flows that harness cells toward body-level anatomical outcomes represent a very attractive and tractable endogenous control system, which is being targeted by emerging approaches to cancer.
Collapse
Affiliation(s)
- Michael Levin
- Allen Discovery Center at Tufts University, 200 Boston Ave., Suite 4600, Medford, MA, 02155, USA.
| |
Collapse
|
17
|
Checchetto V, Leanza L, De Stefani D, Rizzuto R, Gulbins E, Szabo I. Mitochondrial K + channels and their implications for disease mechanisms. Pharmacol Ther 2021; 227:107874. [PMID: 33930454 DOI: 10.1016/j.pharmthera.2021.107874] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
The field of mitochondrial ion channels underwent a rapid development during the last decade, thanks to the molecular identification of some of the nuclear-encoded organelle channels and to advances in strategies allowing specific pharmacological targeting of these proteins. Thereby, genetic tools and specific drugs aided definition of the relevance of several mitochondrial channels both in physiological as well as pathological conditions. Unfortunately, in the case of mitochondrial K+ channels, efforts of genetic manipulation provided only limited results, due to their dual localization to mitochondria and to plasma membrane in most cases. Although the impact of mitochondrial K+ channels on human diseases is still far from being genuinely understood, pre-clinical data strongly argue for their substantial role in the context of several pathologies, including cardiovascular and neurodegenerative diseases as well as cancer. Importantly, these channels are druggable targets, and their in-depth investigation could thus pave the way to the development of innovative small molecules with huge therapeutic potential. In the present review we summarize the available experimental evidence that mechanistically link mitochondrial potassium channels to the above pathologies and underline the possibility of exploiting them for therapy.
Collapse
Affiliation(s)
| | - Luigi Leanza
- Department of Biology, University of Padova, Italy
| | | | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Italy
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Germany
| | - Ildiko Szabo
- Department of Biology, University of Padova, Italy; CNR Institute of Neurosciences, Italy.
| |
Collapse
|
18
|
Altamura C, Greco MR, Carratù MR, Cardone RA, Desaphy JF. Emerging Roles for Ion Channels in Ovarian Cancer: Pathomechanisms and Pharmacological Treatment. Cancers (Basel) 2021; 13:668. [PMID: 33562306 PMCID: PMC7914442 DOI: 10.3390/cancers13040668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Ovarian cancer (OC) is the deadliest gynecologic cancer, due to late diagnosis, development of platinum resistance, and inadequate alternative therapy. It has been demonstrated that membrane ion channels play important roles in cancer processes, including cell proliferation, apoptosis, motility, and invasion. Here, we review the contribution of ion channels in the development and progression of OC, evaluating their potential in clinical management. Increased expression of voltage-gated and epithelial sodium channels has been detected in OC cells and tissues and shown to be involved in cancer proliferation and invasion. Potassium and calcium channels have been found to play a critical role in the control of cell cycle and in the resistance to apoptosis, promoting tumor growth and recurrence. Overexpression of chloride and transient receptor potential channels was found both in vitro and in vivo, supporting their contribution to OC. Furthermore, ion channels have been shown to influence the sensitivity of OC cells to neoplastic drugs, suggesting a critical role in chemotherapy resistance. The study of ion channels expression and function in OC can improve our understanding of pathophysiology and pave the way for identifying ion channels as potential targets for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
| | - Maria Raffaella Greco
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari Aldo Moro, 70125 Bari, Italy;
| | - Maria Rosaria Carratù
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari Aldo Moro, 70125 Bari, Italy;
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (M.R.G.); (M.R.C.); (J.-F.D.)
| |
Collapse
|
19
|
Bachmann M, Rossa A, Antoniazzi G, Biasutto L, Carrer A, Campagnaro M, Leanza L, Gonczi M, Csernoch L, Paradisi C, Mattarei A, Zoratti M, Szabo I. Synthesis and cellular effects of a mitochondria-targeted inhibitor of the two-pore potassium channel TASK-3. Pharmacol Res 2021; 164:105326. [PMID: 33338625 DOI: 10.1016/j.phrs.2020.105326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/03/2020] [Accepted: 11/23/2020] [Indexed: 01/25/2023]
Abstract
The two-pore potassium channel TASK-3 has been shown to localize to both the plasma membrane and the mitochondrial inner membrane. TASK-3 is highly expressed in melanoma and breast cancer cells and has been proposed to promote tumor formation. Here we investigated whether pharmacological modulation of TASK-3, and specifically of mitochondrial TASK-3 (mitoTASK-3), had any effect on cancer cell survival and mitochondrial physiology. A novel, mitochondriotropic version of the specific TASK-3 inhibitor IN-THPP has been synthesized by addition of a positively charged triphenylphosphonium moiety. While IN-THPP was unable to induce apoptosis, mitoIN-THPP decreased survival of breast cancer cells and efficiently killed melanoma lines, which we show to express mitoTASK-3. Cell death was accompanied by mitochondrial membrane depolarization and fragmentation of the mitochondrial network, suggesting a role of the channel in the maintenance of the correct function of this organelle. In accordance, cells treated with mitoIN-THPP became rapidly depleted of mitochondrial ATP which resulted in activation of the AMP-dependent kinase AMPK. Importantly, cell survival was not affected in mouse embryonic fibroblasts and the effect of mitoIN-THPP was less pronounced in human melanoma cells stably knocked down for TASK-3 expression, indicating a certain degree of selectivity of the drug both for pathological cells and for the channel. In addition, mitoIN-THPP inhibited cancer cell migration to a higher extent than IN-THPP in two melanoma cell lines. In summary, our results point to the importance of mitoTASK-3 for melanoma cell survival and migration.
Collapse
Affiliation(s)
| | - Andrea Rossa
- Department of Chemical Sciences, University of Padua, Italy
| | | | - Lucia Biasutto
- CNR Institute of Neuroscience, Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy
| | - Andrea Carrer
- Department of Biology, University of Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy
| | | | - Luigi Leanza
- Department of Biology, University of Padua, Italy
| | - Monika Gonczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Hungary
| | - Laszlo Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Hungary
| | | | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Mario Zoratti
- CNR Institute of Neuroscience, Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padua, Italy; CNR Institute of Neuroscience, Padua, Italy.
| |
Collapse
|
20
|
Maltby CJ, Schofield JPR, Houghton SD, O’Kelly I, Vargas-Caballero M, Deinhardt K, Coldwell MJ. A 5' UTR GGN repeat controls localisation and translation of a potassium leak channel mRNA through G-quadruplex formation. Nucleic Acids Res 2020; 48:9822-9839. [PMID: 32870280 PMCID: PMC7515701 DOI: 10.1093/nar/gkaa699] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/28/2020] [Indexed: 12/18/2022] Open
Abstract
RNA G-quadruplexes (G4s) are secondary structures proposed to function as regulators of post-transcriptional mRNA localisation and translation. G4s within some neuronal mRNAs are known to control distal localisation and local translation, contributing to distinct local proteomes that facilitate the synaptic remodelling attributed to normal cellular function. In this study, we characterise the G4 formation of a (GGN)13 repeat found within the 5' UTR of the potassium 2-pore domain leak channel Task3 mRNA. Biophysical analyses show that this (GGN)13 repeat forms a parallel G4 in vitro exhibiting the stereotypical potassium specificity of G4s, remaining thermostable under physiological ionic conditions. Through mouse brain tissue G4-RNA immunoprecipitation, we further confirm that Task3 mRNA forms a G4 structure in vivo. The G4 is inhibitory to translation of Task3 in vitro and is overcome through activity of a G4-specific helicase DHX36, increasing K+ leak currents and membrane hyperpolarisation in HEK293 cells. Further, we observe that this G4 is fundamental to ensuring delivery of Task3 mRNA to distal primary cortical neurites. It has been shown that aberrant Task3 expression correlates with neuronal dysfunction, we therefore posit that this G4 is important in regulated local expression of Task3 leak channels that maintain K+ leak within neurons.
Collapse
Affiliation(s)
- Connor J Maltby
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - James P R Schofield
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - Steven D Houghton
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - Ita O’Kelly
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | | | - Katrin Deinhardt
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - Mark J Coldwell
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| |
Collapse
|
21
|
Serova OV, Gantsova EA, Deyev IE, Petrenko AG. The Value of pH Sensors in Maintaining Homeostasis of the Nervous System. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162020040196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Pethő Z, Najder K, Carvalho T, McMorrow R, Todesca LM, Rugi M, Bulk E, Chan A, Löwik CWGM, Reshkin SJ, Schwab A. pH-Channeling in Cancer: How pH-Dependence of Cation Channels Shapes Cancer Pathophysiology. Cancers (Basel) 2020; 12:E2484. [PMID: 32887220 PMCID: PMC7565548 DOI: 10.3390/cancers12092484] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue acidosis plays a pivotal role in tumor progression: in particular, interstitial acidosis promotes tumor cell invasion, and is a major contributor to the dysregulation of tumor immunity and tumor stromal cells. The cell membrane and integral membrane proteins commonly act as important sensors and transducers of altered pH. Cell adhesion molecules and cation channels are prominent membrane proteins, the majority of which is regulated by protons. The pathophysiological consequences of proton-sensitive ion channel function in cancer, however, are scarcely considered in the literature. Thus, the main focus of this review is to highlight possible events in tumor progression and tumor immunity where the pH sensitivity of cation channels could be of great importance.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Karolina Najder
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Tiago Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
| | - Luca Matteo Todesca
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Micol Rugi
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Etmar Bulk
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Alan Chan
- Percuros B.V., 2333 CL Leiden, The Netherlands;
| | - Clemens W. G. M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
- Department of Oncology CHUV, UNIL and Ludwig Cancer Center, 1011 Lausanne, Switzerland
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| |
Collapse
|
23
|
Böhme I, Schönherr R, Eberle J, Bosserhoff AK. Membrane Transporters and Channels in Melanoma. Rev Physiol Biochem Pharmacol 2020; 181:269-374. [PMID: 32737752 DOI: 10.1007/112_2020_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent research has revealed that ion channels and transporters can be important players in tumor development, progression, and therapy resistance in melanoma. For example, members of the ABC family were shown to support cancer stemness-like features in melanoma cells, while several members of the TRP channel family were reported to act as tumor suppressors.Also, many transporter proteins support tumor cell viability and thus suppress apoptosis induction by anticancer therapy. Due to the high number of ion channels and transporters and the resulting high complexity of the field, progress in understanding is often focused on single molecules and is in total rather slow. In this review, we aim at giving an overview about a broad subset of ion transporters, also illustrating some aspects of the field, which have not been addressed in detail in melanoma. In context with the other chapters in this special issue on "Transportome Malfunctions in the Cancer Spectrum," a comparison between melanoma and these tumors will be possible.
Collapse
Affiliation(s)
- Ines Böhme
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Schönherr
- Institute of Biochemistry and Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Jürgen Eberle
- Department of Dermatology, Venerology and Allergology, Skin Cancer Center Charité, University Medical Center Charité, Berlin, Germany
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany. .,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
24
|
Mathie A, Veale EL, Cunningham KP, Holden RG, Wright PD. Two-Pore Domain Potassium Channels as Drug Targets: Anesthesia and Beyond. Annu Rev Pharmacol Toxicol 2020; 61:401-420. [PMID: 32679007 DOI: 10.1146/annurev-pharmtox-030920-111536] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Two-pore domain potassium (K2P) channels stabilize the resting membrane potential of both excitable and nonexcitable cells and, as such, are important regulators of cell activity. There are many conditions where pharmacological regulation of K2P channel activity would be of therapeutic benefit, including, but not limited to, atrial fibrillation, respiratory depression, pulmonary hypertension, neuropathic pain, migraine, depression, and some forms of cancer. Up until now, few if any selective pharmacological regulators of K2P channels have been available. However, recent publications of solved structures with small-molecule activators and inhibitors bound to TREK-1, TREK-2, and TASK-1 K2P channels have given insight into the pharmacophore requirements for compound binding to these sites. Together with the increasing availability of a number of novel, active, small-molecule compounds from K2P channel screening programs, these advances have opened up the possibility of rational activator and inhibitor design to selectively target K2P channels.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | - Emma L Veale
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | - Kevin P Cunningham
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - Robyn G Holden
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | | |
Collapse
|
25
|
Withaferin A suppresses breast cancer cell proliferation by inhibition of the two-pore domain potassium (K2P9) channel TASK-3. Biomed Pharmacother 2020; 129:110383. [PMID: 32563149 DOI: 10.1016/j.biopha.2020.110383] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/26/2020] [Accepted: 06/07/2020] [Indexed: 12/30/2022] Open
Abstract
Withaferin A (WFA), a C5,C6-epoxy steroidal lactone isolated from the medicinal plant Withania somnifera (L.) Dunal, inhibits growth of tumor cells in different cancer types. However, the mechanisms underlying the effect of WFA on tumor cells are not fully understood. In the present study, we evaluated the blockade of TASK-3 channels by WFA in TASK-3-expressing HEK-293 cells. Explore if the WFA-mediated TASK-3 blockade can be used as a pharmacological tool to decrease the cell viability in cancer cells. A combination of functional experiments (patch-clamp, gene downregulation, overexpression and pharmacological inhibition) and molecular docking analysis were used to get insights into the mechanism by which the inhibition of TASK-3 by WFA affects the growth and viability of cancer cells. Withaferin A was found to inhibit the activity of TASK-3 channels. The inhibitory effect of Withaferin A on TASK-3 potassium currents was dose-dependent and independent of voltage. Molecular modeling studies identified putative WFA-binding sites in TASK-3 channel involved the channel blockade. In agreements with the molecular modeling predictions, mutation of residues F125 to A (F125A), L197 to V (L197 V) and the double mutant F125A-L197 V markedly decreased the WFA-induced inhibition of TASK-3. Finally, the cytotoxic effect of WFA was tested in MDA-MB-231 human breast cancer cells transfected with TASK-3 or shRNA that decreases TASK-3 expression. Together, our results show that the cytotoxic effect of WFA on fully transformed MDA-MB-231 cells depends on the expression of TASK-3. Herein, we also provide insights into the mechanism of TASK-3 inhibition by WFA.
Collapse
|
26
|
TASK-3 Gene Knockdown Dampens Invasion and Migration and Promotes Apoptosis in KATO III and MKN-45 Human Gastric Adenocarcinoma Cell Lines. Int J Mol Sci 2019; 20:ijms20236077. [PMID: 31810225 PMCID: PMC6928893 DOI: 10.3390/ijms20236077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
Incidence and mortality of gastric cancer is increasing worldwide, in part, because of the lack of new therapeutic targets to treat this disease. Different types of ion channels participate in the hallmarks of cancer. In this context, ion channels are known to exert control over the cell cycle, mechanisms that support survival, angiogenesis, migration, and cell invasion. In particular, TASK-3 (KCNK9), a member of the K2P potassium channel family, has attracted much interest because of its oncogenic properties. However, despite multiple lines of evidence linking TASK-3 to tumorigenesis in various types of cancer, its relationship with gastric cancer has not been fully examined. Therefore, we set out to assess the effect of TASK-3 gene knockdown on KATO III and MKN-45 human gastric adenocarcinoma cell lines by using a short hairpin RNA (shRNA)-mediated knockdown. Our results demonstrate that knocking down TASK-3 reduces cell proliferation and viability because of an increase in apoptosis without an apparent effect on cell cycle checkpoints. In addition, cell migration and invasion are reduced after knocking down TASK-3 in these cell lines. The present study highlights TASK-3 as a key protein involved in migration and cell survival in gastric cancer and corroborates its potential as a therapeutic target for gastric cancer treatment.
Collapse
|
27
|
Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol 2019; 25:5732-5772. [PMID: 31636470 PMCID: PMC6801186 DOI: 10.3748/wjg.v25.i38.5732] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
In their seminal papers Hanahan and Weinberg described oncogenic processes a normal cell undergoes to be transformed into a cancer cell. The functions of ion channels in the gastrointestinal (GI) tract influence a variety of cellular processes, many of which overlap with these hallmarks of cancer. In this review we focus on the roles of the calcium (Ca2+), sodium (Na+), potassium (K+), chloride (Cl-) and zinc (Zn2+) transporters in GI cancer, with a special emphasis on the roles of the KCNQ1 K+ channel and CFTR Cl- channel in colorectal cancer (CRC). Ca2+ is a ubiquitous second messenger, serving as a signaling molecule for a variety of cellular processes such as control of the cell cycle, apoptosis, and migration. Various members of the TRP superfamily, including TRPM8, TRPM7, TRPM6 and TRPM2, have been implicated in GI cancers, especially through overexpression in pancreatic adenocarcinomas and down-regulation in colon cancer. Voltage-gated sodium channels (VGSCs) are classically associated with the initiation and conduction of action potentials in electrically excitable cells such as neurons and muscle cells. The VGSC NaV1.5 is abundantly expressed in human colorectal CRC cell lines as well as being highly expressed in primary CRC samples. Studies have demonstrated that conductance through NaV1.5 contributes significantly to CRC cell invasiveness and cancer progression. Zn2+ transporters of the ZIP/SLC39A and ZnT/SLC30A families are dysregulated in all major GI organ cancers, in particular, ZIP4 up-regulation in pancreatic cancer (PC). More than 70 K+ channel genes, clustered in four families, are found expressed in the GI tract, where they regulate a range of cellular processes, including gastrin secretion in the stomach and anion secretion and fluid balance in the intestinal tract. Several distinct types of K+ channels are found dysregulated in the GI tract. Notable are hERG1 upregulation in PC, gastric cancer (GC) and CRC, leading to enhanced cancer angiogenesis and invasion, and KCNQ1 down-regulation in CRC, where KCNQ1 expression is associated with enhanced disease-free survival in stage II, III, and IV disease. Cl- channels are critical for a range of cellular and tissue processes in the GI tract, especially fluid balance in the colon. Most notable is CFTR, whose deficiency leads to mucus blockage, microbial dysbiosis and inflammation in the intestinal tract. CFTR is a tumor suppressor in several GI cancers. Cystic fibrosis patients are at a significant risk for CRC and low levels of CFTR expression are associated with poor overall disease-free survival in sporadic CRC. Two other classes of chloride channels that are dysregulated in GI cancers are the chloride intracellular channels (CLIC1, 3 & 4) and the chloride channel accessory proteins (CLCA1,2,4). CLIC1 & 4 are upregulated in PC, GC, gallbladder cancer, and CRC, while the CLCA proteins have been reported to be down-regulated in CRC. In summary, it is clear, from the diverse influences of ion channels, that their aberrant expression and/or activity can contribute to malignant transformation and tumor progression. Further, because ion channels are often localized to the plasma membrane and subject to multiple layers of regulation, they represent promising clinical targets for therapeutic intervention including the repurposing of current drugs.
Collapse
Affiliation(s)
- Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| |
Collapse
|
28
|
Riedelsberger J, Obando PA, Gonzalez W. Yeast strain Saccharomyces cerevisiae BYT45 lacking the cation extrusion systems ENA1-5 and NHA1 is suitable for the characterization of TASK-3 potassium channel antagonists. FEMS Yeast Res 2019; 19:5524363. [PMID: 31247642 DOI: 10.1093/femsyr/foz044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 06/24/2019] [Indexed: 11/13/2022] Open
Abstract
Finding new potential antagonists of potassium channels is a continuing task. TASK potassium channels operate over a large physiological range of membrane voltages, why they are thought to contribute to the excitability and resting potential of mammalian membrane potentials. Additionally, they are regulated by extracellular stimuli like changes in pH and K+ concentrations. TASK malfunctions are associated with diseases, which makes them popular targets for the search of new antagonists. Identification of channel inhibitors can be a time-consuming and expensive project. Here, we present an easy-to-use and inexpensive yeast system for the expression of the two-pore domain K+ channel TASK-3, and for the characterization of TASK-3 antagonists. The Saccharomyces cerevisiae strain BYT45 was used to express guinea pig TASK-3. The system allowed the expression and characterization of TASK-3 at variable pH values and K+ concentrations. Three known TASK-3 antagonists have been tested in the BYT45 yeast system: PK-THPP, ZnCl2 and Bupivacaine. Their inhibitory effect on TASK-3 was tested in solid and liquid media assays, and half maximal inhibitory concentrations were estimated. Although the system is less sensitive than more refined systems, the antagonistic activity could be confirmed for all three inhibitors.
Collapse
Affiliation(s)
- Janin Riedelsberger
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 3460000 Talca, Chile
| | - Patricia A Obando
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 3460000 Talca, Chile
| | - Wendy Gonzalez
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 3460000 Talca, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Centro de Bioinformática y Simulación Molecular, Universidad de Talca, 2 Norte 685, 3460000 Talca, Chile
| |
Collapse
|
29
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
30
|
Tuszynski J, Tilli TM, Levin M. Ion Channel and Neurotransmitter Modulators as Electroceutical Approaches to the Control of Cancer. Curr Pharm Des 2019; 23:4827-4841. [PMID: 28554310 PMCID: PMC6340161 DOI: 10.2174/1381612823666170530105837] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 11/22/2022]
Abstract
The activities of individual cells must be tightly coordinated in order to build and maintain complex 3-dimensional body structures during embryogenesis and regeneration. Thus, one way to view cancer is within systems biology as a network disorder affecting the ability of cells to properly interact with a morphodynamic field of instructive signals that keeps proliferation and migration orchestrated toward the anatomical needs of the host or-ganism. One layer of this set of instructive microenvironmental cues is bioelectrical. Voltage gradients among all somatic cells (not just excitable nerve and muscle) control cell behavior, and the ionic coupling of cells into networks via electrochemical synapses allows them to implement tissue-level patterning decisions. These gradients have been increasingly impli-cated in the induction and suppression of tumorigenesis and metastasis, in the emerging links between developmental bioelectricity to the cancer problem. Consistent with the well-known role of neurotransmitter molecules in transducing electrical activity to downstream cascades in the brain, serotonergic signaling has likewise been implicated in cancer. Here, we review these recent data and propose new approaches for manipulating bioelectric and neurotransmitter pathways in cancer biology based on a bioelectric view of cancer. To sup-port this methodology, we present new data on the effects of the SSRI Prozac and its analog (ZINC ID = ZINC06811610) on survival of both cancer (MCF7) and normal (MCF10A) breast cells exposed to these compounds. We found an IC50 concentration (25 μM for Pro-zac and 100 μM for the Prozac analog) at which these compounds inhibited tumor cell sur-vival and proliferation. Additionally, at these concentrations, we did not observe alterations in a non-tumoral cell line. This constitutes a proof-of-concept demonstration for our hy-pothesis that the use of both existing and novel drugs as electroceuticals could serve as an alternative to highly toxic chemotherapy strategies replacing or augmenting them with less toxic alternatives. We believe this new approach forms an exciting roadmap for future bio-medical advances.
Collapse
Affiliation(s)
- Jack Tuszynski
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta. Canada
| | - Tatiana M Tilli
- Laboratory of Biological System Modeling, National Institute for Science and Technology on Innovation in Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro. Brazil
| | - Michael Levin
- Biology Department, and Allen Discovery Center, Tufts University, Medford, MA, 02155. United States
| |
Collapse
|
31
|
Zavala WD, Foscolo MR, Kunda PE, Cavicchia JC, Acosta CG. Changes in the expression of the potassium channels TASK1, TASK3 and TRESK in a rat model of oral squamous cell carcinoma and their relation to malignancy. Arch Oral Biol 2019; 100:75-85. [PMID: 30818127 DOI: 10.1016/j.archoralbio.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Potassium channels have been proposed to promote cancer cell proliferation and metastases. Thus, we investigated the expression pattern of three 2-pore domain potassium channels (K2Ps) TASK1, TASK3 and TRESK in advanced oral squamous cell carcinoma (OSCC), the commonest oral malignancy. DESIGN We used 4-nitroquinoline-1-oxide (4-NQO) to induce high grade OSCC in male adult rats. We then used immunohistochemistry and Western blotting to study the distribution and expression pattern of TASK1, TASK3 and TRESK in normal versus cancerous tissue. We also examined the expression of β-tubulin III (β-tub3), a marker associated with resistance to taxane-based chemotherapy and poor patient prognosis, and its correlation with the K2Ps. Finally, we studied the expression of TASK1, TASK3 and TRESK in human samples of SCC of oral origin. RESULTS We found that TASK3 was significantly up-regulated whereas TASK1 and TRESK were both significantly down-regulated in advanced, poorly differentiated OSCC. Both, rat and human SCC showed a significant increase in the expression of β-tub3. Interestingly, the expression of the latter correlated positively and significantly with TASK3 and TRESK but not TASK1 in rat OSCC. Our initial results showed a similar pattern of up and down regulation and correlation with β-tub3 for these three K2Ps in human SCC. CONCLUSIONS The changes in expression and the co-localization with a marker of resistance to taxanes like β-tub3 turn TASK1, TASK3 and TRESK into potentially new prognostic tools and possibly new therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Walther D Zavala
- Facultad de Odontología, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Mabel R Foscolo
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Patricia E Kunda
- Centro Investigación Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina.
| | - Juan C Cavicchia
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Cristian G Acosta
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
32
|
Prevarskaya N, Skryma R, Shuba Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol Rev 2018; 98:559-621. [PMID: 29412049 DOI: 10.1152/physrev.00044.2016] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Genomic instability is a primary cause and fundamental feature of human cancer. However, all cancer cell genotypes generally translate into several common pathophysiological features, often referred to as cancer hallmarks. Although nowadays the catalog of cancer hallmarks is quite broad, the most common and obvious of them are 1) uncontrolled proliferation, 2) resistance to programmed cell death (apoptosis), 3) tissue invasion and metastasis, and 4) sustained angiogenesis. Among the genes affected by cancer, those encoding ion channels are present. Membrane proteins responsible for signaling within cell and among cells, for coupling of extracellular events with intracellular responses, and for maintaining intracellular ionic homeostasis ion channels contribute to various extents to pathophysiological features of each cancer hallmark. Moreover, tight association of these hallmarks with ion channel dysfunction gives a good reason to classify them as special type of channelopathies, namely oncochannelopathies. Although the relation of cancer hallmarks to ion channel dysfunction differs from classical definition of channelopathies, as disease states causally linked with inherited mutations of ion channel genes that alter channel's biophysical properties, in a broader context of the disease state, to which pathogenesis ion channels essentially contribute, such classification seems absolutely appropriate. In this review the authors provide arguments to substantiate such point of view.
Collapse
Affiliation(s)
- Natalia Prevarskaya
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Roman Skryma
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Yaroslav Shuba
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| |
Collapse
|
33
|
Bachmann M, Costa R, Peruzzo R, Prosdocimi E, Checchetto V, Leanza L. Targeting Mitochondrial Ion Channels to Fight Cancer. Int J Mol Sci 2018; 19:ijms19072060. [PMID: 30011966 PMCID: PMC6073807 DOI: 10.3390/ijms19072060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, several experimental evidences have underlined a new role of ion channels in cancer development and progression. In particular, mitochondrial ion channels are arising as new oncological targets, since it has been proved that most of them show an altered expression during tumor development and the pharmacological targeting of some of them have been demonstrated to be able to modulate cancer growth and progression, both in vitro as well as in vivo in pre-clinical mouse models. In this scenario, pharmacology of mitochondrial ion channels would be in the near future a new frontier for the treatment of tumors. In this review, we discuss the new advances in the field, by focusing our attention on the improvements in new drug developments to target mitochondrial ion channels.
Collapse
Affiliation(s)
| | - Roberto Costa
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | - Roberta Peruzzo
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | - Elena Prosdocimi
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | | | - Luigi Leanza
- Department of Biology, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
34
|
|
35
|
Zúñiga R, Valenzuela C, Concha G, Brown N, Zúñiga L. TASK-3 Downregulation Triggers Cellular Senescence and Growth Inhibition in Breast Cancer Cell Lines. Int J Mol Sci 2018; 19:ijms19041033. [PMID: 29596383 PMCID: PMC5979529 DOI: 10.3390/ijms19041033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/28/2022] Open
Abstract
TASK-3 potassium channels are believed to promote proliferation and survival of cancer cells, in part, by augmenting their resistance to both hypoxia and serum deprivation. While overexpression of TASK-3 is frequently observed in cancers, the understanding of its role and regulation during tumorigenesis remains incomplete. Here, we evaluated the effect of reducing the expression of TASK-3 in MDA-MB-231 and MCF-10F human mammary epithelial cell lines through small hairpin RNA (shRNA)-mediated knockdown. Our results show that knocking down TASK-3 in fully transformed MDA-MB-231 cells reduces proliferation, which was accompanied by an induction of cellular senescence and cell cycle arrest, with an upregulation of cyclin-dependent kinase (CDK) inhibitors p21 and p27. In non-tumorigenic MCF-10F cells, however, TASK-3 downregulation did not lead to senescence induction, although cell proliferation was impaired and an upregulation of CDK inhibitors was also evident. Our observations implicate TASK-3 as a critical factor in cell cycle progression and corroborate its potential as a therapeutic target in breast cancer treatment.
Collapse
Affiliation(s)
- Rafael Zúñiga
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Claudio Valenzuela
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Guierdy Concha
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Nelson Brown
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Leandro Zúñiga
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| |
Collapse
|
36
|
Moore D, Walker SI, Levin M. Cancer as a disorder of patterning information: computational and biophysical perspectives on the cancer problem. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa8548] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Dookeran KA, Zhang W, Stayner L, Argos M. Associations of two-pore domain potassium channels and triple negative breast cancer subtype in The Cancer Genome Atlas: systematic evaluation of gene expression and methylation. BMC Res Notes 2017; 10:475. [PMID: 28899398 PMCID: PMC5596847 DOI: 10.1186/s13104-017-2777-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Objectives It is unclear whether 2-pore domain potassium channels are novel molecular markers with differential expression related to biologically aggressive triple-negative type breast tumors. Our objective was to systematically evaluate associations of 2-pore domain potassium channel gene expression and DNA methylation with triple-negative subtype in The Cancer Genome Atlas invasive breast cancer dataset. Methylation and expression data for all fifteen 2-pore domain potassium family genes were examined for 1040 women, and associations with triple-negative subtype (vs. luminal A) were evaluated using age/race adjusted generalized-linear models, with Bonferroni-corrected significance thresholds. Subtype associated CpG loci were evaluated for functionality related to expression using Spearman’s correlation. Results Overexpression of KCNK5, KCNK9 and KCNK12, and underexpression of KCNK6 and KCNK15, were significantly associated with triple-negative subtype (Bonferroni-corrected p < 0.0033). A total of 195 (114 hypomethylated and 81 hypermethylated) CpG loci were found to be significantly associated with triple-negative subtype (Bonferroni-corrected p < 8.22 × 10−8). Significantly negatively correlated expression patterns that were differentially observed in triple-negative vs. luminal A subtype were demonstrated for: KCNK2 (gene body: cg04923840, cg13916421), KCNK5 (gene body: cg05255811, cg18705155, cg09130674, cg21388745, cg00859574) and KCNK9 (TSS1500: cg21415530, cg12175729; KCNK9/TRAPPC9 intergenic region: cg17336929, cg25900813, cg03919980). CpG loci listed for KCNK5 and KCNK9 all showed relative hypomethylation for probability of triple-negative vs. luminal A subtype. Triple-negative subtype was associated with distinct 2-pore domain potassium channel expression patterns. Both KCNK5 and KCNK9 overexpression appeared to be functionally related to CpG loci hypomethylation. Electronic supplementary material The online version of this article (doi:10.1186/s13104-017-2777-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Keith A Dookeran
- Epidemiology, University of Wisconsin-Milwaukee, Joseph J. Zilber School of Public Health, 1240 N. 10th St, Milwaukee, WI, 53205, USA. .,The Cancer Foundation for Minority and Underserved Populations, Chicago, IL, 60614, USA.
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Drive, Suite 1400, Chicago, IL, 60611, USA
| | - Leslie Stayner
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, School of Public Health, 1603 West Taylor Street, MC923, Chicago, IL, 60612, USA
| | - Maria Argos
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, School of Public Health, 1603 West Taylor Street, MC923, Chicago, IL, 60612, USA
| |
Collapse
|
38
|
Bohnen MS, Roman-Campos D, Terrenoire C, Jnani J, Sampson KJ, Chung WK, Kass RS. The Impact of Heterozygous KCNK3 Mutations Associated With Pulmonary Arterial Hypertension on Channel Function and Pharmacological Recovery. J Am Heart Assoc 2017; 6:JAHA.117.006465. [PMID: 28889099 PMCID: PMC5634293 DOI: 10.1161/jaha.117.006465] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Heterozygous loss of function mutations in the KCNK3 gene cause hereditary pulmonary arterial hypertension (PAH). KCNK3 encodes an acid-sensitive potassium channel, which contributes to the resting potential of human pulmonary artery smooth muscle cells. KCNK3 is widely expressed in the body, and dimerizes with other KCNK3 subunits, or the closely related, acid-sensitive KCNK9 channel. METHODS AND RESULTS We engineered homomeric and heterodimeric mutant and nonmutant KCNK3 channels associated with PAH. Using whole-cell patch-clamp electrophysiology in human pulmonary artery smooth muscle and COS7 cell lines, we determined that homomeric and heterodimeric mutant channels in heterozygous KCNK3 conditions lead to mutation-specific severity of channel dysfunction. Both wildtype and mutant KCNK3 channels were activated by ONO-RS-082 (10 μmol/L), causing cell hyperpolarization. We observed robust gene expression of KCNK3 in healthy and familial PAH patient lungs, but no quantifiable expression of KCNK9, and demonstrated in functional studies that KCNK9 minimizes the impact of select KCNK3 mutations when the 2 channel subunits co-assemble. CONCLUSIONS Heterozygous KCNK3 mutations in PAH lead to variable loss of channel function via distinct mechanisms. Homomeric and heterodimeric mutant KCNK3 channels represent novel therapeutic substrates in PAH. Pharmacological and pH-dependent activation of wildtype and mutant KCNK3 channels in pulmonary artery smooth muscle cells leads to membrane hyperpolarization. Co-assembly of KCNK3 with KCNK9 subunits may provide protection against KCNK3 loss of function in tissues where both KCNK9 and KCNK3 are expressed, contributing to the lung-specific phenotype observed clinically in patients with PAH because of KCNK3 mutations.
Collapse
Affiliation(s)
- Michael S Bohnen
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| | | | - Cecile Terrenoire
- Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Jack Jnani
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Kevin J Sampson
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Wendy K Chung
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Robert S Kass
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
39
|
Kim HK, Noh YH, Nilius B, Ko KS, Rhee BD, Kim N, Han J. Current and upcoming mitochondrial targets for cancer therapy. Semin Cancer Biol 2017. [PMID: 28627410 DOI: 10.1016/j.semcancer.2017.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria are essential intracellular organelles that regulate energy metabolism, cell death, and signaling pathways that are important for cell proliferation and differentiation. Therefore, mitochondria are fundamentally implicated in cancer biology, including initiation, growth, metastasis, relapse, and acquired drug resistance. Based on these implications, mitochondria have been proposed as a major therapeutic target for cancer treatment. In addition to classical view of mitochondria in cancer biology, recent studies found novel pathophysiological roles of mitochondria in cancer. In this review, we introduce recent concepts of mitochondrial roles in cancer biology including mitochondrial DNA mutation and epigenetic modulation, energy metabolism reprogramming, mitochondrial channels, involvement in metastasis and drug resistance, and cancer stem cells. We also discuss the role of mitochondria in emerging cancer therapeutic strategies, especially cancer immunotherapy and CRISPR-Cas9 system gene therapy.
Collapse
Affiliation(s)
- Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea; Department of Integrated Biomedical Science, College of Medicine, Inje University, Busan, Republic of Korea
| | - Yeon Hee Noh
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Bernd Nilius
- KU Leuven, Department Cell Mol Medicine, Leuven, 3000, Belgium
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea.
| |
Collapse
|
40
|
Shi C, Thum C, Zhang Q, Tu W, Pelaz B, Parak WJ, Zhang Y, Schneider M. Inhibition of the cancer-associated TASK 3 channels by magnetically induced thermal release of Tetrandrine from a polymeric drug carrier. J Control Release 2016; 237:50-60. [DOI: 10.1016/j.jconrel.2016.06.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/09/2016] [Accepted: 06/29/2016] [Indexed: 12/15/2022]
|
41
|
Rezania S, Kammerer S, Li C, Steinecker-Frohnwieser B, Gorischek A, DeVaney TTJ, Verheyen S, Passegger CA, Tabrizi-Wizsy NG, Hackl H, Platzer D, Zarnani AH, Malle E, Jahn SW, Bauernhofer T, Schreibmayer W. Overexpression of KCNJ3 gene splice variants affects vital parameters of the malignant breast cancer cell line MCF-7 in an opposing manner. BMC Cancer 2016; 16:628. [PMID: 27519272 PMCID: PMC4983040 DOI: 10.1186/s12885-016-2664-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/03/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Overexpression the KCNJ3, a gene that encodes subunit 1 of G-protein activated inwardly rectifying K(+) channel (GIRK1) in the primary tumor has been found to be associated with reduced survival times and increased lymph node metastasis in breast cancer patients. METHODS In order to survey possible tumorigenic properties of GIRK1 overexpression, a range of malignant mammary epithelial cells, based on the MCF-7 cell line that permanently overexpress different splice variants of the KCNJ3 gene (GIRK1a, GIRK1c, GIRK1d and as a control, eYFP) were produced. Subsequently, selected cardinal neoplasia associated cellular parameters were assessed and compared. RESULTS Adhesion to fibronectin coated surface as well as cell proliferation remained unaffected. Other vital parameters intimately linked to malignancy, i.e. wound healing, chemoinvasion, cellular velocities / motilities and angiogenesis were massively affected by GIRK1 overexpression. Overexpression of different GIRK1 splice variants exerted differential actions. While GIRK1a and GIRK1c overexpression reinforced the affected parameters towards malignancy, overexpression of GIRK1d resulted in the opposite. Single channel recording using the patch clamp technique revealed functional GIRK channels in the plasma membrane of MCF-7 cells albeit at very low frequency. DISCUSSION We conclude that GIRK1d acts as a dominant negative constituent of functional GIRK complexes present in the plasma membrane of MCF-7 cells, while overexpression of GIRK1a and GIRK1c augmented their activity. The core component responsible for the cancerogenic action of GIRK1 is apparently presented by a segment comprising aminoacids 235-402, that is present exclusively in GIRK1a and GIRK1c, but not GIRK1d (positions according to GIRK1a primary structure). CONCLUSIONS The current study provides insight into the cellular and molecular consequences of KCNJ3 overexpression in breast cancer cells and the mechanism upon clinical outcome in patients suffering from breast cancer.
Collapse
Affiliation(s)
- S. Rezania
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| | - S. Kammerer
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| | - C. Li
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| | - B. Steinecker-Frohnwieser
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
- Present address: Institute of Human Genetics, Medical University of Graz, Graz, Austria
| | - A. Gorischek
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| | - T. T. J. DeVaney
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| | - S. Verheyen
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
- Present address: Institute of Human Genetics, Medical University of Graz, Graz, Austria
| | - C. A. Passegger
- Institute of Pathophysiology and Immunology, SFL Chicken CAM Laboratory, Medical University of Graz, Graz, Austria
| | - N. Ghaffari Tabrizi-Wizsy
- Institute of Pathophysiology and Immunology, SFL Chicken CAM Laboratory, Medical University of Graz, Graz, Austria
| | - H. Hackl
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - D. Platzer
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
| | - A. H. Zarnani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - E. Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - S. W. Jahn
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - T. Bauernhofer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| | - W. Schreibmayer
- Institute of Biophysics, Molecular Physiology Group, Medical University of Graz, Harrachgasse 21/4, Graz, Austria
- Research Unit on Ion Channels and Cancer Biology, Medical University of Graz, Graz, Austria
| |
Collapse
|
42
|
TASK-1 Regulates Apoptosis and Proliferation in a Subset of Non-Small Cell Lung Cancers. PLoS One 2016; 11:e0157453. [PMID: 27294516 PMCID: PMC4905626 DOI: 10.1371/journal.pone.0157453] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/31/2016] [Indexed: 12/29/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide; survival times are poor despite therapy. The role of the two-pore domain K+ (K2P) channel TASK-1 (KCNK3) in lung cancer is at present unknown. We found that TASK-1 is expressed in non-small cell lung cancer (NSCLC) cell lines at variable levels. In a highly TASK-1 expressing NSCLC cell line, A549, a characteristic pH- and hypoxia-sensitive non-inactivating K+ current was measured, indicating the presence of functional TASK-1 channels. Inhibition of TASK-1 led to significant depolarization in these cells. Knockdown of TASK-1 by siRNA significantly enhanced apoptosis and reduced proliferation in A549 cells, but not in weakly TASK-1 expressing NCI-H358 cells. Na+-coupled nutrient transport across the cell membrane is functionally coupled to the efflux of K+ via K+ channels, thus TASK-1 may potentially influence Na+-coupled nutrient transport. In contrast to TASK-1, which was not differentially expressed in lung cancer vs. normal lung tissue, we found the Na+-coupled nutrient transporters, SLC5A3, SLC5A6, and SLC38A1, transporters for myo-inositol, biotin and glutamine, respectively, to be significantly overexpressed in lung adenocarcinomas. In summary, we show for the first time that the TASK-1 channel regulates apoptosis and proliferation in a subset of NSCLC.
Collapse
|
43
|
Pharmacological targeting of ion channels for cancer therapy: In vivo evidences. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1385-97. [DOI: 10.1016/j.bbamcr.2015.11.032] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022]
|
44
|
Sun H, Luo L, Lal B, Ma X, Chen L, Hann CL, Fulton AM, Leahy DJ, Laterra J, Li M. A monoclonal antibody against KCNK9 K(+) channel extracellular domain inhibits tumour growth and metastasis. Nat Commun 2016; 7:10339. [PMID: 26842342 PMCID: PMC4742836 DOI: 10.1038/ncomms10339] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 11/30/2015] [Indexed: 12/25/2022] Open
Abstract
Two-pore domain potassium (K2P) channels act to maintain cell resting membrane potential--a prerequisite for many biological processes. KCNK9, a member of K2P family, is implicated in cancer, owing to its overexpression in human tumours and its ability to promote neoplastic cell survival and growth. However, KCNK9's underlying contributions to malignancy remain elusive due to the absence of specific modulators. Here we describe the development of monoclonal antibodies against the KCNK9 extracellular domain and their functional effects. We show that one antibody (Y4) with the highest affinity binding induces channel internalization. The addition of Y4 to KCNK9-expressing carcinoma cells reduces cell viability and increases cell death. Systemic administration of Y4 effectively inhibits growth of human lung cancer xenografts and murine breast cancer metastasis in mice. Evidence for Y4-mediated carcinoma cell autonomous and immune-dependent cytotoxicity is presented. Our study reveals that antibody-based KCNK9 targeting is a promising therapeutic strategy in KCNK9-expressing malignancies.
Collapse
Affiliation(s)
- Han Sun
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205, USA
| | - Liqun Luo
- Immunotherapy Institute, Fujian Medical University, Fujian 350108, China
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205, USA
| | - Xinrong Ma
- Department of Pathology, University of Maryland, Baltimore, Maryland 21201, USA
| | - Lieping Chen
- Department of Immunobiology and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Christine L Hann
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Amy M Fulton
- Department of Pathology, University of Maryland, Baltimore, Maryland 21201, USA.,Baltimore Veterans Administration Medical Center, Baltimore, Maryland 21201, USA
| | - Daniel J Leahy
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - John Laterra
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Min Li
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
45
|
Vierra NC, Dadi PK, Jeong I, Dickerson M, Powell DR, Jacobson DA. Type 2 Diabetes-Associated K+ Channel TALK-1 Modulates β-Cell Electrical Excitability, Second-Phase Insulin Secretion, and Glucose Homeostasis. Diabetes 2015; 64:3818-28. [PMID: 26239056 PMCID: PMC4613978 DOI: 10.2337/db15-0280] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/22/2015] [Indexed: 12/11/2022]
Abstract
Two-pore domain K+ (K2P) channels play an important role in tuning β-cell glucose-stimulated insulin secretion (GSIS). The K2P channel TWIK-related alkaline pH-activated K2P (TALK)-1 is linked to type 2 diabetes risk through a coding sequence polymorphism (rs1535500); however, its physiological function has remained elusive. Here, we show that TALK-1 channels are expressed in mouse and human β-cells, where they serve as key regulators of electrical excitability and GSIS. We find that the rs1535500 polymorphism, which results in an alanine-to-glutamate substitution in the C-terminus of human TALK-1, increases channel activity. Genetic ablation of TALK-1 results in β-cell membrane potential depolarization, increased islet Ca2+ influx, and enhanced second-phase GSIS. Moreover, mice lacking TALK-1 channels are resistant to high-fat diet-induced elevations in fasting glycemia. These findings reveal TALK-1 channels as important modulators of second-phase insulin secretion and suggest a clinically relevant mechanism for rs1535500, which may increase type 2 diabetes risk by limiting GSIS.
Collapse
Affiliation(s)
- Nicholas C Vierra
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Prasanna K Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Imju Jeong
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Matthew Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | | | - David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
46
|
Aghazadeh Y, Papadopoulos V. The role of the 14-3-3 protein family in health, disease, and drug development. Drug Discov Today 2015; 21:278-87. [PMID: 26456530 DOI: 10.1016/j.drudis.2015.09.012] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 09/21/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022]
Abstract
14-3-3 proteins regulate intracellular signaling pathways, such as signal transduction, protein trafficking, cell cycle, and apoptosis. In addition to the ubiquitous roles of 14-3-3 isoforms, unique tissue-specific functions are also described for each isoform. Owing to their role in regulating cell cycle, protein trafficking, and steroidogenesis, 14-3-3 proteins are prevalent in human diseases, such as cancer, neurodegeneration, and reproductive disorders, and, therefore, serve as valuable drug targets. In this review, we summarize the role of 14-3-3 proteins in normal and disease states, with a focus on 14-3-3γ and ɛ. We also discuss drug compounds targeting 14-3-3 proteins and their potential therapeutic uses.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
47
|
Levin M. Molecular bioelectricity: how endogenous voltage potentials control cell behavior and instruct pattern regulation in vivo. Mol Biol Cell 2015; 25:3835-50. [PMID: 25425556 PMCID: PMC4244194 DOI: 10.1091/mbc.e13-12-0708] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In addition to biochemical gradients and transcriptional networks, cell behavior is regulated by endogenous bioelectrical cues originating in the activity of ion channels and pumps, operating in a wide variety of cell types. Instructive signals mediated by changes in resting potential control proliferation, differentiation, cell shape, and apoptosis of stem, progenitor, and somatic cells. Of importance, however, cells are regulated not only by their own Vmem but also by the Vmem of their neighbors, forming networks via electrical synapses known as gap junctions. Spatiotemporal changes in Vmem distribution among nonneural somatic tissues regulate pattern formation and serve as signals that trigger limb regeneration, induce eye formation, set polarity of whole-body anatomical axes, and orchestrate craniofacial patterning. New tools for tracking and functionally altering Vmem gradients in vivo have identified novel roles for bioelectrical signaling and revealed the molecular pathways by which Vmem changes are transduced into cascades of downstream gene expression. Because channels and gap junctions are gated posttranslationally, bioelectrical networks have their own characteristic dynamics that do not reduce to molecular profiling of channel expression (although they couple functionally to transcriptional networks). The recent data provide an exciting opportunity to crack the bioelectric code, and learn to program cellular activity at the level of organs, not only cell types. The understanding of how patterning information is encoded in bioelectrical networks, which may require concepts from computational neuroscience, will have transformative implications for embryogenesis, regeneration, cancer, and synthetic bioengineering.
Collapse
Affiliation(s)
- Michael Levin
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155-4243
| |
Collapse
|
48
|
Renigunta V, Schlichthörl G, Daut J. Much more than a leak: structure and function of K₂p-channels. Pflugers Arch 2015; 467:867-94. [PMID: 25791628 DOI: 10.1007/s00424-015-1703-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 11/27/2022]
Abstract
Over the last decade, we have seen an enormous increase in the number of experimental studies on two-pore-domain potassium channels (K2P-channels). The collection of reviews and original articles compiled for this special issue of Pflügers Archiv aims to give an up-to-date summary of what is known about the physiology and pathophysiology of K2P-channels. This introductory overview briefly describes the structure of K2P-channels and their function in different organs. Its main aim is to provide some background information for the 19 reviews and original articles of this special issue of Pflügers Archiv. It is not intended to be a comprehensive review; instead, this introductory overview focuses on some unresolved questions and controversial issues, such as: Do K2P-channels display voltage-dependent gating? Do K2P-channels contribute to the generation of action potentials? What is the functional role of alternative translation initiation? Do K2P-channels have one or two or more gates? We come to the conclusion that we are just beginning to understand the extremely complex regulation of these fascinating channels, which are often inadequately described as 'leak channels'.
Collapse
Affiliation(s)
- Vijay Renigunta
- Institute of Physiology and Pathophysiology, Marburg University, 35037, Marburg, Germany
| | | | | |
Collapse
|
49
|
Chernet BT, Levin M. Transmembrane voltage potential of somatic cells controls oncogene-mediated tumorigenesis at long-range. Oncotarget 2015; 5:3287-306. [PMID: 24830454 PMCID: PMC4102810 DOI: 10.18632/oncotarget.1935] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The microenvironment is increasingly recognized as a crucial aspect of cancer. In contrast and complement to the field's focus on biochemical factors and extracellular matrix, we characterize a novel aspect of host:tumor interaction - endogenous bioelectric signals among non-excitable somatic cells. Extending prior work focused on the bioelectric state of cancer cells themselves, we show for the first time that the resting potentials of distant cells are critical for oncogene-dependent tumorigenesis. In the Xenopus laevis tadpole model, we used human oncogenes such as mutant KRAS to drive formation of tumor-like structures that exhibited overproliferation, increased nuclear size, hypoxia, acidity, and leukocyte attraction. Remarkably, misexpression of hyperpolarizing ion channels at distant sites within the tadpole significantly reduced the incidence of these tumors. The suppression of tumorigenesis could also be achieved by hyperpolarization using native CLIC1 chloride channels, suggesting a treatment modality not requiring gene therapy. Using a dominant negative approach, we implicate HDAC1 as the mechanism by which resting potential changes affect downstream cell behaviors. Based on published data on the voltage-mediated changes of butyrate flux through the SLC5A8 transporter, we present a model linking resting potentials of host cells to the ability of oncogenes to initiate tumorigenesis. Antibiotic data suggest that the relevant butyrate is generated by a native bacterial species, identifying a novel link between the microbiome and cancer that is mediated by alterations in bioelectric signaling.
Collapse
Affiliation(s)
- Brook T Chernet
- Center for Regenerative and Developmental Biology and Department of Biology Tufts University 200 Boston Avenue,Suite 4600 Medford, MA 02155 U.S.A
| | | |
Collapse
|
50
|
Feliciangeli S, Chatelain FC, Bichet D, Lesage F. The family of K2P channels: salient structural and functional properties. J Physiol 2015; 593:2587-603. [PMID: 25530075 DOI: 10.1113/jphysiol.2014.287268] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/10/2014] [Indexed: 12/11/2022] Open
Abstract
Potassium channels participate in many biological functions, from ion homeostasis to generation and modulation of the electrical membrane potential. They are involved in a large variety of diseases. In the human genome, 15 genes code for K(+) channels with two pore domains (K2P ). These channels form dimers of pore-forming subunits that produce background conductances finely regulated by a range of natural and chemical effectors, including signalling lipids, temperature, pressure, pH, antidepressants and volatile anaesthetics. Since the cloning of TWIK1, the prototypical member of this family, a lot of work has been carried out on their structure and biology. These studies are still in progress, but data gathered so far show that K2P channels are central players in many processes, including ion homeostasis, hormone secretion, cell development and excitability. A growing number of studies underline their implication in physiopathological mechanisms, such as vascular and pulmonary hypertension, cardiac arrhythmias, nociception, neuroprotection and depression. This review gives a synthetic view of the most noticeable features of these channels.
Collapse
Affiliation(s)
- Sylvain Feliciangeli
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Frank C Chatelain
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Delphine Bichet
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Florian Lesage
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| |
Collapse
|