201
|
Moreels L, Peigneur S, Yamaguchi Y, Vriens K, Waelkens E, Zhu S, Thevissen K, Cammue BPA, Sato K, Tytgat J. Expanding the pharmacological profile of κ-hefutoxin 1 and analogues: A focus on the inhibitory effect on the oncogenic channel K v10.1. Peptides 2017; 98:43-50. [PMID: 27578329 DOI: 10.1016/j.peptides.2016.08.008] [Citation(s) in RCA: 16] [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: 05/27/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 10/21/2022]
Abstract
Peptide toxins, such as scorpion peptides, are interesting lead compounds in the search for novel drugs. In this paper, the focus is on the scorpion peptide κ-hefutoxin 1. This peptide displays a cysteine-stabilized helix-loop-helix fold (CSα/α) and is known to be a weak Kv1.x inhibitor. Due to the low affinity of κ-hefutoxin 1 for these channels, it is assumed that the main target(s) of κ-hefutoxin 1 remain(s) unknown. In order to identify novel targets, electrophysiological measurements and antifungal assays were performed. The effect of κ-hefutoxin 1 was previously evaluated on a panel of 11 different voltage-gated potassium channels. Here, we extended this target screening with the oncogenic potassium channel Kv10.1. κ-Hefutoxin 1 was able to inhibit this channel in a dose-dependent manner (IC50∼26μM). Although the affinity is rather low, this is the first peptide toxin ever described to be a Kv10.1 inhibitor. The structure-activity relationship of κ-hefutoxin 1 on Kv10.1 was investigated by testing eight κ-hefutoxin 1 variants using the two-electrode voltage clamp technique. Several important amino acid residues were identified; the functional dyad residues (Tyr5 and Lys19), N-terminal residues (Gly1 and His2) and the amidated C-terminal residue (Cys22). Since the CSα/α fold is also found in a class of antifungal plant peptides, the α-hairpinines, we investigated the antifungal activity of κ-hefutoxin 1. κ-Hefutoxin 1 showed low activity against the plant pathogen Fusarium culmorum and no activity against three other yeast and fungal species, even at high concentrations (∼100μM).
Collapse
Affiliation(s)
- Lien Moreels
- Toxicology and Pharmacology, KU Leuven, Campus Gasthuisberg O&N2, Herestraat 49, PO Box 922, 3000 Leuven, Belgium.
| | - Steve Peigneur
- Toxicology and Pharmacology, KU Leuven, Campus Gasthuisberg O&N2, Herestraat 49, PO Box 922, 3000 Leuven, Belgium.
| | - Yoko Yamaguchi
- Department of Environmental Science, Fukuoka Women's University, Fukuoka 813-8529, Japan.
| | - Kim Vriens
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, PO Box 2460, 3001 Leuven, Belgium.
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Campus Gasthuisberg, O&N1, Herestraat 49, PO Box 901, 3000 Leuven, Belgium.
| | - Shunyi Zhu
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects & Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, PO Box 2460, 3001 Leuven, Belgium.
| | - Bruno P A Cammue
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, PO Box 2460, 3001 Leuven, Belgium; VIB Department of Plant Systems Biology, Technologiepark 927, 9052 Ghent, Belgium.
| | - Kazuki Sato
- Department of Environmental Science, Fukuoka Women's University, Fukuoka 813-8529, Japan.
| | - Jan Tytgat
- Toxicology and Pharmacology, KU Leuven, Campus Gasthuisberg O&N2, Herestraat 49, PO Box 922, 3000 Leuven, Belgium.
| |
Collapse
|
202
|
Stankevich N, Mosekilde E. Coexistence between silent and bursting states in a biophysical Hodgkin-Huxley-type of model. CHAOS (WOODBURY, N.Y.) 2017; 27:123101. [PMID: 29289049 DOI: 10.1063/1.4986401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Classification of the dynamical mechanisms that support bistability between bursting oscillations and silence has not yet been clarified in detail. The purpose of this paper is to demonstrate that the coexistence of a stable equilibrium point with a state of continuous bursting can occur in a slightly modified, biophysical model that describe the dynamics of pancreatic beta-cells. To realize this form of coexistence, we have introduced an additional voltage-dependent potassium current that is activated in the region around the original, unstable equilibrium point. It is interesting to note that this modification also leads the model to display a blue-sky catastrophe in the transition region between chaotic and bursting states.
Collapse
Affiliation(s)
- Nataliya Stankevich
- Department of Radio-Electronics and Telecommunications, Yuri Gagarin State Technical University of Saratov, 77, Politechnicheskaya, Saratov 410054, Russian Federation
| | - Erik Mosekilde
- Department of Physics, Technical University of Denmark, 2800 Lyngby, Denmark
| |
Collapse
|
203
|
The combined activation of K Ca3.1 and inhibition of K v11.1/hERG1 currents contribute to overcome Cisplatin resistance in colorectal cancer cells. Br J Cancer 2017; 118:200-212. [PMID: 29161243 PMCID: PMC5785745 DOI: 10.1038/bjc.2017.392] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Platinum-based drugs such as Cisplatin are commonly employed for cancer treatment. Despite an initial therapeutic response, Cisplatin treatment often results in the development of chemoresistance. To identify novel approaches to overcome Cisplatin resistance, we tested Cisplatin in combination with K+ channel modulators on colorectal cancer (CRC) cells. Methods: The functional expression of Ca2+-activated (KCa3.1, also known as KCNN4) and voltage-dependent (Kv11.1, also known as KCNH2 or hERG1) K+ channels was determined in two CRC cell lines (HCT-116 and HCT-8) by molecular and electrophysiological techniques. Cisplatin and several K+ channel modulators were tested in vitro for their action on K+ currents, cell vitality, apoptosis, cell cycle, proliferation, intracellular signalling and Platinum uptake. These effects were also analysed in a mouse model mimicking Cisplatin resistance. Results: Cisplatin-resistant CRC cells expressed higher levels of KCa3.1 and Kv11.1 channels, compared with Cisplatin-sensitive CRC cells. In resistant cells, KCa3.1 activators (SKA-31) and Kv11.1 inhibitors (E4031) had a synergistic action with Cisplatin in triggering apoptosis and inhibiting proliferation. The effect was maximal when KCa3.1 activation and Kv11.1 inhibition were combined. In fact, similar results were produced by Riluzole, which is able to both activate KCa3.1 and inhibit Kv11.1. Cisplatin uptake into resistant cells depended on KCa3.1 channel activity, as it was potentiated by KCa3.1 activators. Kv11.1 blockade led to increased KCa3.1 expression and thereby stimulated Cisplatin uptake. Finally, the combined administration of a KCa3.1 activator and a Kv11.1 inhibitor also overcame Cisplatin resistance in vivo. Conclusions: As Riluzole, an activator of KCa3.1 and inhibitor of Kv11.1 channels, is in clinical use, our results suggest that this compound may be useful in the clinic to improve Cisplatin efficacy and overcome Cisplatin resistance in CRC.
Collapse
|
204
|
Mendoza PA, Silva P, Díaz J, Arriagada C, Canales J, Cerda O, Torres VA. Calpain2 mediates Rab5-driven focal adhesion disassembly and cell migration. Cell Adh Migr 2017; 12:185-194. [PMID: 29099266 DOI: 10.1080/19336918.2017.1377388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The early endosome protein Rab5 was recently shown to promote cell migration by enhancing focal adhesion disassembly through mechanisms that remain elusive. Focal adhesion disassembly is associated to proteolysis of talin, in a process that requires calpain2. Since calpain2 has been found at vesicles and endosomal compartments, we hypothesized that Rab5 stimulates calpain2 activity, leading to enhanced focal adhesion disassembly in migrating cells. We observed that calpain2 co-localizes with EEA1-positive early endosomes and co-immunoprecipitates with EEA1 and Rab5 in A549 lung carcinoma cells undergoing spreading, whereas Rab5 knock-down decreased the accumulation of calpain2 at early endosomal-enriched fractions. In addition, Rab5 silencing decreased calpain2 activity, as shown by cleavage of the fluorogenic substrate tBOC-LM-CMAC and the endogenous substrate talin. Accordingly, Rab5 promoted focal adhesion disassembly in a calpain2-dependent manner, as expression of GFP-Rab5 accelerated focal adhesion disassembly in nocodazole-synchronized cells, whereas pharmacological inhibition of calpain2 with N-acetyl-Leu-Leu-Met prevented both focal adhesion disassembly and cell migration induced by Rab5. In summary, these data uncover Rab5 as a novel regulator of calpain2 activity and focal adhesion proteolysis leading to cell migration.
Collapse
Affiliation(s)
- Pablo A Mendoza
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,b Molecular Pathology Laboratory , Institute of Biochemistry and Microbiology, Sciences Faculty, Universidad Austral de Chile , Valdivia , Chile
| | - Patricio Silva
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,c Faculty of Health Sciences, Universidad Central de Chile , Santiago , Chile
| | - Jorge Díaz
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,d Advanced Center for Chronic Diseases (ACCDiS) , Universidad de Chile , Santiago , Chile
| | - Cecilia Arriagada
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile
| | - Jimena Canales
- e Programa de Biología Celular y Molecular , Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Oscar Cerda
- e Programa de Biología Celular y Molecular , Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile , Santiago , Chile.,f Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Chile , Santiago , Chile
| | - Vicente A Torres
- a Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile , Santiago , Chile.,d Advanced Center for Chronic Diseases (ACCDiS) , Universidad de Chile , Santiago , Chile
| |
Collapse
|
205
|
Herrera F, Sevrain CM, Jaffrès PA, Couthon H, Grélard A, Dufourc EJ, Chantôme A, Potier-Cartereau M, Vandier C, Bouchet AM. Singular Interaction between an Antimetastatic Agent and the Lipid Bilayer: The Ohmline Case. ACS OMEGA 2017; 2:6361-6370. [PMID: 30023517 PMCID: PMC6045331 DOI: 10.1021/acsomega.7b00936] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/08/2017] [Indexed: 06/08/2023]
Abstract
SK3 channels are abnormaly expressed in metastatic cells, and Ohmline (OHM), an ether lipid, has been shown to reduce the activity of SK3 channels and the migration capacity of cancer cells. OHM incorporation into the plasma membrane is proposed to dissociate the protein complex formed between SK3 and Orai1, a potassium and a calcium channel, respectively, and would lead to a modification in the lipid environment of both the proteins. Here, we report the synthesis of deuterated OHM that affords the determination, through solid-state NMR, of its entire partitioning into membranes mimicking the SK3 environment. Use of deuterated lipids affords the demonstration of an OHM-induced membrane disordering, which is dose-dependent and increases with increasing amounts of cholesterol (CHOL). Molecular dynamics simulations comfort the disordering action and show that OHM interacts with the carbonyl and phosphate groups of stearoylphosphatidylcholine and sphingomyelin and to a minor extent with CHOL. OHM is thus proposed to remove the CHOL OH moieties away from their main binding sites, forcing a new rearrangement with other lipid groups. Such an interaction takes its origin at the lipid-water interface, but it propagates toward the entire lipid molecules and leads to a cooperative destabilization of the lipid acyl chains, that is, membrane disordering. The consequences of this reorganization of the lipid phases are discussed in the context of the OHM-induced inhibition of SK3 channels.
Collapse
Affiliation(s)
- Fernando
E. Herrera
- Physics
Department, Universidad Nacional del Litoral,
Ciudad Universitaria, 3000 Santa Fe, Argentina
| | - Charlotte M. Sevrain
- Université
de Brest, CEMCA, UMR CNRS 6521, IBSAM, 6, Avenue Victor le Gorgeu, 29238 Brest, France
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
| | - Paul-Alain Jaffrès
- Université
de Brest, CEMCA, UMR CNRS 6521, IBSAM, 6, Avenue Victor le Gorgeu, 29238 Brest, France
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
| | - Hélène Couthon
- Université
de Brest, CEMCA, UMR CNRS 6521, IBSAM, 6, Avenue Victor le Gorgeu, 29238 Brest, France
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
| | - Axelle Grélard
- Université
Bordeaux, Institute of Chemistry & Biology of Membranes &
Nanoobjects, UMR5248 CNRS, Allée de Geoffroy St Hilaire Bât B14 Pessac, 33600 Bordeaux, France
| | - Erick J. Dufourc
- Université
Bordeaux, Institute of Chemistry & Biology of Membranes &
Nanoobjects, UMR5248 CNRS, Allée de Geoffroy St Hilaire Bât B14 Pessac, 33600 Bordeaux, France
| | - Aurélie Chantôme
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
- Université
François Rabelais de Tours, Nutrition, Croissance et Cancer, Inserm UMR1069, 10 Boulevard Tonnellé Bât. Dutrochet, 2ème étage, 37032 Tours, France
| | - Marie Potier-Cartereau
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
- Université
François Rabelais de Tours, Nutrition, Croissance et Cancer, Inserm UMR1069, 10 Boulevard Tonnellé Bât. Dutrochet, 2ème étage, 37032 Tours, France
| | - Christophe Vandier
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
- Université
François Rabelais de Tours, Nutrition, Croissance et Cancer, Inserm UMR1069, 10 Boulevard Tonnellé Bât. Dutrochet, 2ème étage, 37032 Tours, France
| | - Ana M. Bouchet
- Network
and Cancer-Canceropole Grand Ouest, (IC-CGO), Maison de la Recherche
en Santé, 63 Quai
Magellan, 44000 Nantes, France
- Université
François Rabelais de Tours, Nutrition, Croissance et Cancer, Inserm UMR1069, 10 Boulevard Tonnellé Bât. Dutrochet, 2ème étage, 37032 Tours, France
| |
Collapse
|
206
|
Steudel FA, Mohr CJ, Stegen B, Nguyen HY, Barnert A, Steinle M, Beer‐Hammer S, Koch P, Lo W, Schroth W, Hoppe R, Brauch H, Ruth P, Huber SM, Lukowski R. SK4 channels modulate Ca 2+ signalling and cell cycle progression in murine breast cancer. Mol Oncol 2017; 11:1172-1188. [PMID: 28557306 PMCID: PMC5579333 DOI: 10.1002/1878-0261.12087] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/27/2017] [Accepted: 05/16/2017] [Indexed: 01/08/2023] Open
Abstract
Oncogenic signalling via Ca2+ -activated K+ channels of intermediate conductance (SK4, also known as KCa 3.1 or IK) has been implicated in different cancer entities including breast cancer. Yet, the role of endogenous SK4 channels for tumorigenesis is unclear. Herein, we generated SK4-negative tumours by crossing SK4-deficient (SK4 KO) mice to the polyoma middle T-antigen (PyMT) and epidermal growth factor receptor 2 (cNeu) breast cancer models in which oncogene expression is driven by the retroviral promoter MMTV. Survival parameters and tumour progression were studied in cancer-prone SK4 KO in comparison with wild-type (WT) mice and in a syngeneic orthotopic mouse model following transplantation of SK4-negative or WT tumour cells. SK4 activity was modulated by genetic or pharmacological means using the SK4 inhibitor TRAM-34 in order to establish the role of breast tumour SK4 for cell growth, electrophysiological signalling, and [Ca2+ ]i oscillations. Ablation of SK4 and TRAM-34 treatment reduced the SK4-generated current fraction, growth factor-dependent Ca2+ entry, cell cycle progression and the proliferation rate of MMTV-PyMT tumour cells. In vivo, PyMT oncogene-driven tumorigenesis was only marginally affected by the global lack of SK4, whereas tumour progression was significantly delayed after orthotopic implantation of MMTV-PyMT SK4 KO breast tumour cells. However, overall survival and progression-free survival time in the MMTV-cNeu mouse model were significantly extended in the absence of SK4. Collectively, our data from murine breast cancer models indicate that SK4 activity is crucial for cell cycle control. Thus, the modulation of this channel should be further investigated towards a potential improvement of existing antitumour strategies in human breast cancer.
Collapse
Affiliation(s)
- Friederike A. Steudel
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
| | - Corinna J. Mohr
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgart and University of TuebingenGermany
| | - Benjamin Stegen
- Department of Radiation OncologyUniversity of TuebingenGermany
| | - Hoang Y. Nguyen
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
| | - Andrea Barnert
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
| | - Marc Steinle
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
| | - Sandra Beer‐Hammer
- Department of Pharmacology and Experimental TherapyInstitute of Experimental and Clinical Pharmacology and ToxicologyUniversity Hospital TuebingenGermany
| | - Pierre Koch
- Pharmaceutical and Medicinal ChemistryInstitute of PharmacyUniversity of TuebingenGermany
| | - Wing‐Yee Lo
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgart and University of TuebingenGermany
| | - Werner Schroth
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgart and University of TuebingenGermany
| | - Reiner Hoppe
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgart and University of TuebingenGermany
| | - Hiltrud Brauch
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgart and University of TuebingenGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
| | | | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical PharmacyInstitute of PharmacyUniversity of TuebingenGermany
| |
Collapse
|
207
|
Wang L, He J, Xia A, Cheng M, Yang Q, Du C, Wei H, Huang X, Zhou Q. Toxic effects of environmental rare earth elements on delayed outward potassium channels and their mechanisms from a microscopic perspective. CHEMOSPHERE 2017; 181:690-698. [PMID: 28476009 DOI: 10.1016/j.chemosphere.2017.04.141] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 06/07/2023]
Abstract
The wide applications cause a large amount of rare earth elements (REEs) to be released into the environment, and ultimately into the human body through food chain. Toxic effects of REEs on humans have been extensively studied, but their toxic effects and binding targets in cells are not understood. Delayed outward potassium channels (K+ channels) are good targets for exogenous substances or clinical drugs. To evaluate cellular toxicities of REEs and clarify toxic mechanisms, the toxicities of REEs on the K+ channel and their structural basis were investigated. The results showed that delayed outward potassium channels on the plasma membrane are the targets of REEs acting on living organisms, and the changes in the thermodynamic and kinetic characteristics of the K+ channel are the reasons of diseases induced by REEs. Two types of REEs, a light REE La3+ and a heavy REE Tb3+, displayed different intensity of toxicities on the K+ channel, in which the toxicity of Tb3+ was stronger than that of La3+. More interestingly, in comparison with that of heavy metal Cd2+, the cytotoxicities of the light and heavy REEs showed discriminative differences, and the cytotoxicity of Tb3+ was higher than that of Cd2+, while the cytotoxicity of La3+ was lower than that of Cd2+. These different cytotoxicities of La3+, Tb3+ and Cd2+ on human resulted from the varying binding abilities of the metals to this channel protein.
Collapse
Affiliation(s)
- Lihong Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China; State Key Laboratory of Food Science and Technology, Jiangsu Key Laboratory of Anaerobic Biotechnology, School of Environment and Civil Engineering, Jiangnan University, Wuxi 214122, China
| | - Jingfang He
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Ao Xia
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Mengzhu Cheng
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Qing Yang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Chunlei Du
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Haiyan Wei
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Xiaohua Huang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China.
| | - Qing Zhou
- State Key Laboratory of Food Science and Technology, Jiangsu Key Laboratory of Anaerobic Biotechnology, School of Environment and Civil Engineering, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
208
|
Lu FF, Wang HY, He XZ, Liang TY, Wang W, Hu HM, Wu F, Liu YW, Zhang SZ. Prognostic value of ion channel genes in Chinese patients with gliomas based on mRNA expression profiling. J Neurooncol 2017; 134:397-405. [DOI: 10.1007/s11060-017-2539-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 06/25/2017] [Indexed: 01/17/2023]
|
209
|
Bandulik S. Of channels and pumps: different ways to boost the aldosterone? Acta Physiol (Oxf) 2017; 220:332-360. [PMID: 27862984 DOI: 10.1111/apha.12832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/10/2016] [Accepted: 11/11/2016] [Indexed: 01/19/2023]
Abstract
The mineralocorticoid aldosterone is a major factor controlling the salt and water balance and thereby also the arterial blood pressure. Accordingly, primary aldosteronism (PA) characterized by an inappropriately high aldosterone secretion is the most common form of secondary hypertension. The physiological stimulation of aldosterone synthesis in adrenocortical glomerulosa cells by angiotensin II and an increased plasma K+ concentration depends on a membrane depolarization and an increase in the cytosolic Ca2+ activity. Recurrent gain-of-function mutations of ion channels and transporters have been identified in a majority of cases of aldosterone-producing adenomas and in familial forms of PA. In this review, the physiological role of these genes in the regulation of aldosterone synthesis and the altered function of the mutant proteins as well are described. The specific changes of the membrane potential and the cellular ion homoeostasis in adrenal cells expressing the different mutants are compared, and their impact on autonomous aldosterone production and proliferation is discussed.
Collapse
Affiliation(s)
- S. Bandulik
- Medical Cell Biology; University of Regensburg; Regensburg Germany
| |
Collapse
|
210
|
Thurber AE, Nelson M, Frost CL, Levin M, Brackenbury WJ, Kaplan DL. IK channel activation increases tumor growth and induces differential behavioral responses in two breast epithelial cell lines. Oncotarget 2017; 8:42382-42397. [PMID: 28415575 PMCID: PMC5522074 DOI: 10.18632/oncotarget.16389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 03/08/2017] [Indexed: 12/26/2022] Open
Abstract
Many potassium channel families are over-expressed in cancer, but their mechanistic role in disease progression is poorly understood. Potassium channels modulate membrane potential (Vmem) and thereby influence calcium ion dynamics and other voltage-sensitive signaling mechanisms, potentially acting as transcriptional regulators. This study investigated the differential response to over-expression and activation of a cancer-associated potassium channel, the intermediate conductance calcium-activated potassium channel (IK), on aggressive behaviors in mammary epithelial and breast cancer cell lines. IK was over-expressed in the highly metastatic breast cancer cell line MDA-MB-231 and the spontaneously immortalized breast epithelial cell line MCF-10A, and the effect on cancer-associated behaviors was assessed. IK over-expression increased primary tumor growth and metastasis of MDA-MB-231 in orthotopic xenografts, demonstrating for the first time in any cancer type that increased IK is sufficient to promote cancer aggression. The primary tumors had similar vascularization as determined by CD31 staining and similar histological characteristics. Interestingly, despite the increased in vivo growth and metastasis, neither IK over-expression nor activation with agonist had a significant effect on MDA-MB-231 proliferation, invasion, or migration in vitro. In contrast, IK decreased MCF-10A proliferation and invasion through Matrigel but had no effect on migration in a scratch-wound assay. We conclude that IK activity is sufficient to promote cell aggression in vivo. Our data provide novel evidence supporting IK and downstream signaling networks as potential targets for cancer therapies.
Collapse
Affiliation(s)
- Amy E. Thurber
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Michaela Nelson
- Department of Biology, University of York, Heslington, York, UK
| | | | - Michael Levin
- Biology Department, and Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, USA
| | | | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
211
|
Fortunato A. The role of hERG1 ion channels in epithelial-mesenchymal transition and the capacity of riluzole to reduce cisplatin resistance in colorectal cancer cells. Cell Oncol (Dordr) 2017; 40:367-378. [DOI: 10.1007/s13402-017-0328-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2017] [Indexed: 01/08/2023] Open
|
212
|
Nagaraja S, Vitanza NA, Woo PJ, Taylor KR, Liu F, Zhang L, Li M, Meng W, Ponnuswami A, Sun W, Ma J, Hulleman E, Swigut T, Wysocka J, Tang Y, Monje M. Transcriptional Dependencies in Diffuse Intrinsic Pontine Glioma. Cancer Cell 2017; 31:635-652.e6. [PMID: 28434841 PMCID: PMC5462626 DOI: 10.1016/j.ccell.2017.03.011] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/27/2016] [Accepted: 03/22/2017] [Indexed: 12/12/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric cancer with limited therapeutic options. The majority of cases of DIPG exhibit a mutation in histone-3 (H3K27M) that results in oncogenic transcriptional aberrancies. We show here that DIPG is vulnerable to transcriptional disruption using bromodomain inhibition or CDK7 blockade. Targeting oncogenic transcription through either of these methods synergizes with HDAC inhibition, and DIPG cells resistant to HDAC inhibitor therapy retain sensitivity to CDK7 blockade. Identification of super-enhancers in DIPG provides insights toward the cell of origin, highlighting oligodendroglial lineage genes, and reveals unexpected mechanisms mediating tumor viability and invasion, including potassium channel function and EPH receptor signaling. The findings presented demonstrate transcriptional vulnerabilities and elucidate previously unknown mechanisms of DIPG pathobiology.
Collapse
Affiliation(s)
- Surya Nagaraja
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | | | - Pamelyn J Woo
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Kathryn R Taylor
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Fang Liu
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China
| | - Lei Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China
| | - Meng Li
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China
| | - Wei Meng
- Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Anitha Ponnuswami
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Wenchao Sun
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Esther Hulleman
- Department of Pediatric Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Tomek Swigut
- Department of Chemical and Systems Biology, Stanford University, Palo Alto, CA 94305, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University, Palo Alto, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California 94305, USA; Department of Developmental Biology, Stanford University, Palo Alto, California 94305, USA; Howard Hughes Medical Institute, Stanford School of Medicine, Stanford University, Palo Alto, California 94305, USA
| | - Yujie Tang
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA; Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China; Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| | - Michelle Monje
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California 94305, USA.
| |
Collapse
|
213
|
Knockout of the LRRC26 subunit reveals a primary role of LRRC26-containing BK channels in secretory epithelial cells. Proc Natl Acad Sci U S A 2017; 114:E3739-E3747. [PMID: 28416688 DOI: 10.1073/pnas.1703081114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Leucine-rich-repeat-containing protein 26 (LRRC26) is the regulatory γ1 subunit of Ca2+- and voltage-dependent BK-type K+ channels. BK channels that contain LRRC26 subunits are active near normal resting potentials even without Ca2+, suggesting they play unique physiological roles, likely limited to very specific cell types and cellular functions. By using Lrrc26 KO mice with a β-gal reporter, Lrrc26 promoter activity is found in secretory epithelial cells, especially acinar epithelial cells in lacrimal and salivary glands, and also goblet and Paneth cells in intestine and colon, although absent from neurons. We establish the presence of LRRC26 protein in eight secretory tissues or tissues with significant secretory epithelium and show that LRRC26 protein coassembles with the pore-forming BK α-subunit in at least three tissues: lacrimal gland, parotid gland, and colon. In lacrimal, parotid, and submandibular gland acinar cells, LRRC26 KO shifts BK gating to be like α-subunit-only BK channels. Finally, LRRC26 KO mimics the effect of SLO1/BK KO in reducing [K+] in saliva. LRRC26-containing BK channels are competent to contribute to resting K+ efflux at normal cell membrane potentials with resting cytosolic Ca2+ concentrations and likely play a critical physiological role in supporting normal secretory function in all secretory epithelial cells.
Collapse
|
214
|
Bidirectional KCNQ1:β-catenin interaction drives colorectal cancer cell differentiation. Proc Natl Acad Sci U S A 2017; 114:4159-4164. [PMID: 28373572 DOI: 10.1073/pnas.1702913114] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The K+ channel KCNQ1 has been proposed as a tumor suppressor in colorectal cancer (CRC). We investigated the molecular mechanisms regulating KCNQ1:β-catenin bidirectional interactions and their effects on CRC differentiation, proliferation, and invasion. Molecular and pharmacologic approaches were used to determine the influence of KCNQ1 expression on the Wnt/β-catenin signaling and epithelial-to-mesenchymal transition (EMT) in human CRC cell lines of varying stages of differentiation. The expression of KCNQ1 was lost with increasing mesenchymal phenotype in poorly differentiated CRC cell lines as a consequence of repression of the KCNQ1 promoter by β-catenin:T-cell factor (TCF)-4. In well-differentiated epithelial CRC cell lines, KCNQ1 was localized to the plasma membrane in a complex with β-catenin and E-cadherin. The colocalization of KCNQ1 with adherens junction proteins was lost with increasing EMT phenotype. ShRNA knock-down of KCNQ1 caused a relocalization of β-catenin from the plasma membrane and a loss of epithelial phenotype in CRC spheroids. Overexpression of KCNQ1 trapped β-catenin at the plasma membrane, induced a patent lumen in CRC spheroids, and slowed CRC cell invasion. The KCNQ1 ion channel inhibitor chromanol 293B caused membrane depolarization, redistribution of β-catenin into the cytosol, and a reduced transepithelial electrical resistance, and stimulated CRC cell proliferation. Analysis of human primary CRC tumor patient databases showed a positive correlation between KCNQ1:KCNE3 channel complex expression and disease-free survival. We conclude that the KCNQ1 ion channel is a target gene and regulator of the Wnt/β-catenin pathway, and its repression leads to CRC cell proliferation, EMT, and tumorigenesis.
Collapse
|
215
|
Rybarczyk P, Vanlaeys A, Brassart B, Dhennin-Duthille I, Chatelain D, Sevestre H, Ouadid-Ahidouch H, Gautier M. The Transient Receptor Potential Melastatin 7 Channel Regulates Pancreatic Cancer Cell Invasion through the Hsp90α/uPA/MMP2 pathway. Neoplasia 2017; 19:288-300. [PMID: 28284058 PMCID: PMC5345960 DOI: 10.1016/j.neo.2017.01.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/13/2017] [Indexed: 12/28/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a very poor prognosis. There is an urgent need to better understand the molecular mechanisms that regulate PDAC cell aggressiveness. The transient receptor potential melastatin 7 (TRPM7) is a nonselective cationic channel that mainly conducts Ca2+ and Mg2+. TRPM7 is overexpressed in numerous malignancies including PDAC. In the present study, we used the PANC-1 and MIA PaCa-2 cell lines to specifically assess the role of TRPM7 in cell invasion and matrix metalloproteinase secretion. We show that TRPM7 regulates Mg2+ homeostasis and constitutive cation entry in both PDAC cell lines. Moreover, cell invasion is strongly reduced by TRPM7 silencing without affecting the cell viability. Conditioned media were further studied, by gel zymography, to detect matrix metalloproteinase (MMP) secretion in PDAC cells. Our results show that MMP-2, urokinase plasminogen activator (uPA), and heat-shock protein 90α (Hsp90α) secretions are significantly decreased in TRPM7-deficient PDAC cells. Moreover, TRPM7 expression in human PDAC lymph node metastasis is correlated to the channel expression in primary tumor. Taken together, our results show that TRPM7 is involved in PDAC cell invasion through regulation of Hsp90α/uPA/MMP-2 proteolytic axis, confirming that this channel could be a promising biomarker and possibly a target for PDAC metastasis therapy.
Collapse
Affiliation(s)
- Pierre Rybarczyk
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Alison Vanlaeys
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Bertrand Brassart
- SFR CAP-Santé (FED 4231); UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), F-51095 Reims, France
| | - Isabelle Dhennin-Duthille
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Denis Chatelain
- Service d'anatomie pathologique, CHU d'Amiens, Université de Picardie Jules Verne, F-80000 Amiens, France, France
| | - Henri Sevestre
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231); Service d'anatomie pathologique, CHU d'Amiens, Université de Picardie Jules Verne, F-80000 Amiens, France, France
| | - Halima Ouadid-Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231)
| | - Mathieu Gautier
- Laboratoire de Physiologie Cellulaire et Moléculaire-EA4667, UFR Sciences, Université de Picardie Jules Verne, F-80039 Amiens, France; SFR CAP-Santé (FED 4231).
| |
Collapse
|
216
|
Xia J, Wang H, Li S, Wu Q, Sun L, Huang H, Zeng M. Ion channels or aquaporins as novel molecular targets in gastric cancer. Mol Cancer 2017; 16:54. [PMID: 28264681 PMCID: PMC5338097 DOI: 10.1186/s12943-017-0622-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer (GC) is a common disease with few effective treatment choices and poor prognosis, and has the second-highest mortality rates among all cancers worldwide. Dysregulation and/or malfunction of ion channels or aquaporins (AQPs) are common in various human cancers. Furthermore, ion channels are involved in numerous important aspects of the tumor aggressive phonotype, such as proliferation, cell cycle, apoptosis, motility, migration, and invasion. Indeed, by localizing in the plasma membrane, ion channels or AQPs can sense and respond to extracellular environment changes; thus, they play a crucial role in cell signaling and cancer progression. These findings have expanded a new area of pharmaceutical exploration for various types of cancer, including GC. The involvement of multiple ion channels, such as voltage-gated potassium and sodium channels, intracellular chloride channels, ‘transient receptor potential’ channels, and AQPs, which have been shown to facilitate the pathogenesis of other tumors, also plays a role in GC. In this review, an overview of ion channel and aquaporin expression and function in carcinogenesis of GC is presented. Studies of ion channels or AQPs will advance our understanding of the molecular genesis of GC and may identify novel and effective targets for the clinical application of GC.
Collapse
Affiliation(s)
- Jianling Xia
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Hospital of the University of Electronic Science and Technology of China, The Western First Round Road, Section 2#32, Chengdu, 610072, China.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongqiang Wang
- Department of Oncology, Zhoushan Hospital, Zhoushan, 316000, China.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shi Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Qinghui Wu
- Department of Urology, Hainan Provincial People's Hospital, Haikou, 570311, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongxiang Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ming Zeng
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Hospital of the University of Electronic Science and Technology of China, The Western First Round Road, Section 2#32, Chengdu, 610072, China.
| |
Collapse
|
217
|
Kuzmenkov AI, Peigneur S, Chugunov AO, Tabakmakher VM, Efremov RG, Tytgat J, Grishin EV, Vassilevski AA. C-Terminal residues in small potassium channel blockers OdK1 and OSK3 from scorpion venom fine-tune the selectivity. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:465-472. [PMID: 28179135 DOI: 10.1016/j.bbapap.2017.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 01/10/2017] [Accepted: 02/03/2017] [Indexed: 01/11/2023]
Abstract
We report isolation, sequencing, and electrophysiological characterization of OSK3 (α-KTx 8.8 in Kalium and Uniprot databases), a potassium channel blocker from the scorpion Orthochirus scrobiculosus venom. Using the voltage clamp technique, OSK3 was tested on a wide panel of 11 voltage-gated potassium channels expressed in Xenopus oocytes, and was found to potently inhibit Kv1.2 and Kv1.3 with IC50 values of ~331nM and ~503nM, respectively. OdK1 produced by the scorpion Odontobuthus doriae differs by just two C-terminal residues from OSK3, but shows marked preference to Kv1.2. Based on the charybdotoxin-potassium channel complex crystal structure, a model was built to explain the role of the variable residues in OdK1 and OSK3 selectivity.
Collapse
Affiliation(s)
- Alexey I Kuzmenkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Biological Faculty, Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven, Leuven 3000, Belgium
| | - Anton O Chugunov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Valentin M Tabakmakher
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven, Leuven 3000, Belgium
| | - Eugene V Grishin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Alexander A Vassilevski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Biological Faculty, Lomonosov Moscow State University, Moscow 119992, Russia.
| |
Collapse
|
218
|
Khatun A, Fujimoto M, Kito H, Niwa S, Suzuki T, Ohya S. Down-Regulation of Ca 2+-Activated K⁺ Channel K Ca1.1 in Human Breast Cancer MDA-MB-453 Cells Treated with Vitamin D Receptor Agonists. Int J Mol Sci 2016; 17:ijms17122083. [PMID: 27973439 PMCID: PMC5187883 DOI: 10.3390/ijms17122083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/01/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022] Open
Abstract
Vitamin D (VD) reduces the risk of breast cancer and improves disease prognoses. Potential VD analogs are being developed as therapeutic agents for breast cancer treatments. The large-conductance Ca2+-activated K+ channel KCa1.1 regulates intracellular Ca2+ signaling pathways and is associated with high grade tumors and poor prognoses. In the present study, we examined the effects of treatments with VD receptor (VDR) agonists on the expression and activity of KCa1.1 in human breast cancer MDA-MB-453 cells using real-time PCR, Western blotting, flow cytometry, and voltage-sensitive dye imaging. Treatments with VDR agonists for 72 h markedly decreased the expression levels of KCa1.1 transcripts and proteins in MDA-MB-453 cells, resulting in the significant inhibition of depolarization responses induced by paxilline, a specific KCa1.1 blocker. The specific proteasome inhibitor MG132 suppressed VDR agonist-induced decreases in KCa1.1 protein expression. These results suggest that KCa1.1 is a new downstream target of VDR signaling and the down-regulation of KCa1.1 through the transcriptional repression of KCa1.1 and enhancement of KCa1.1 protein degradation contribute, at least partly, to the antiproliferative effects of VDR agonists in breast cancer cells.
Collapse
Affiliation(s)
- Anowara Khatun
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Mayu Fujimoto
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Satomi Niwa
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Takayoshi Suzuki
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 403-8334, Japan.
| | - Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| |
Collapse
|
219
|
Oprisan B, Stoica I, Avadanei MI. Morphological changes induced in erythrocyte membrane by the antiepileptic treatment: An atomic force microscopy study. Microsc Res Tech 2016; 80:364-373. [DOI: 10.1002/jemt.22804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/04/2016] [Accepted: 10/22/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Bogdan Oprisan
- Faculty of Medicine, Discipline Biophysics and Medical Physics; “Grigore T. Popa” University of Medicine and Pharmacy; Iasi Romania
| | - Iuliana Stoica
- Department of Polymer Materials Physics; “Petru Poni” Institute of Macromolecular Chemistry; Iasi Romania
| | - Mihaela Iuliana Avadanei
- Department of Polymer Materials Physics; “Petru Poni” Institute of Macromolecular Chemistry; Iasi Romania
| |
Collapse
|
220
|
The interplay between genetic and bioelectrical signaling permits a spatial regionalisation of membrane potentials in model multicellular ensembles. Sci Rep 2016; 6:35201. [PMID: 27731412 PMCID: PMC5059667 DOI: 10.1038/srep35201] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/26/2016] [Indexed: 12/12/2022] Open
Abstract
The single cell-centred approach emphasises ion channels as specific proteins that determine individual properties, disregarding their contribution to multicellular outcomes. We simulate the interplay between genetic and bioelectrical signals in non-excitable cells from the local single-cell level to the long range multicellular ensemble. The single-cell genetic regulation is based on mean-field kinetic equations involving the mRNA and protein concentrations. The transcription rate factor is assumed to depend on the absolute value of the cell potential, which is dictated by the voltage-gated cell ion channels and the intercellular gap junctions. The interplay between genetic and electrical signals may allow translating single-cell states into multicellular states which provide spatio-temporal information. The model results have clear implications for biological processes: (i) bioelectric signals can override slightly different genetic pre-patterns; (ii) ensembles of cells initially at the same potential can undergo an electrical regionalisation because of persistent genetic differences between adjacent spatial regions; and (iii) shifts in the normal cell electrical balance could trigger significant changes in the genetic regulation.
Collapse
|
221
|
Zaccagnino A, Pilarsky C, Tawfik D, Sebens S, Trauzold A, Novak I, Schwab A, Kalthoff H. In silico analysis of the transportome in human pancreatic ductal adenocarcinoma. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:749-763. [PMID: 27652669 DOI: 10.1007/s00249-016-1171-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/18/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022]
Abstract
The altered expression and/or activity of ion channels and transporters (transportome) have been associated with malignant behavior of cancer cells and were proposed to be a hallmark of cancer. However, the impact of altered transportome in epithelial cancers, such as pancreatic ductal adenocarcinoma (PDAC), as well as its pathophysiological consequences, still remains unclear. Here, we report the in silico analysis of 840 transportome genes in PDAC patients' tissues. Our study was focused on the transportome changes and their correlation with functional and behavioral responses in PDAC tumor and stromal compartments. The dysregulated gene expression datasets were filtered using a cut-off of fold-change values ≤-2 or ≥2 (adjusted p value ≤0.05). The dysregulated transportome genes were clearly associated with impaired physiological secretory mechanisms and/or pH regulation, control of cell volume, and cell polarity. Additionally, some down-regulated transportome genes were found to be closely linked to epithelial cell differentiation. Furthermore, the observed decrease in genes coding for calcium and chloride transport might be a mechanism for evasion of apoptosis. In conclusion, the current work provides a comprehensive overview of the altered transportome expression and its association with predicted PDAC malignancy with special focus on the epithelial compartment.
Collapse
Affiliation(s)
- A Zaccagnino
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| | - C Pilarsky
- Department of Surgery, University Clinic, Krankenhausstr. 12, 91054, Erlangen, Germany
| | - D Tawfik
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - S Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - A Trauzold
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - I Novak
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - A Schwab
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27 b, 48149, Muenster, Germany
| | - H Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| |
Collapse
|
222
|
Pointer KB, Clark PA, Eliceiri KW, Salamat MS, Robertson GA, Kuo JS. Administration of Non-Torsadogenic human Ether-à-go-go-Related Gene Inhibitors Is Associated with Better Survival for High hERG-Expressing Glioblastoma Patients. Clin Cancer Res 2016; 23:73-80. [PMID: 27635088 DOI: 10.1158/1078-0432.ccr-15-3169] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 08/12/2016] [Accepted: 09/03/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE Glioblastoma is the most malignant primary brain tumor, with a median survival of less than 2 years. More effective therapeutic approaches are needed to improve clinical outcomes. EXPERIMENTAL DESIGN Glioblastoma patient-derived cells (GPDC) were isolated from patient glioblastomas and implanted in mice to form xenografts. IHC was performed for human Ether-à-go-go-Related Gene (hERG) expression and tumor proliferation. Sphere-forming assays with the hERG blocker E-4031 were performed on a high and low hERG-expressing lines. A glioblastoma tissue microarray (TMA; 115 patients) was used to correlate hERG expression with patient survival. Clinical data were analyzed to determine whether patient survival was affected by incidental administration of hERG inhibitory drugs and the correlative effect of patient glioblastoma hERG expression levels. RESULTS hERG expression was upregulated in glioblastoma xenografts with higher proliferative indices. High hERG-expressing GPDCs showed a reduction in sphere formation when treated with hERG inhibitors compared with low hERG-expressing GPDCs. Glioblastoma TMA analysis showed worse survival for glioblastoma patients with high hERG expression versus low expression-43.5 weeks versus 60.9 weeks, respectively (P = 0.022). Furthermore, patients who received at least one hERG blocker had a better survival rate compared with patients who did not (P = 0.0015). Subgroup analysis showed that glioblastoma patients with high hERG expression who received hERG blockers had improved survival (P = 0.0458). There was no difference in survival for low hERG-expressing glioblastoma patients who received hERG blockers (P = 0.4136). CONCLUSIONS Our findings suggest that hERG is a potential glioblastoma survival marker, and that already approved drugs with non-torsadogenic hERG inhibitory activity may potentially be repurposed as adjuvant glioblastoma therapy in high hERG-expressing glioblastoma patients. Clin Cancer Res; 23(1); 73-80. ©2016 AACRSee related commentary by Arcangeli and Becchetti, p. 3.
Collapse
Affiliation(s)
- Kelli B Pointer
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul A Clark
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - M Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Gail A Robertson
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Cardiovascular Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - John S Kuo
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Surgery, National University of Singapore, Singapore
| |
Collapse
|
223
|
Șterbuleac D, Maniu CL. An antiarrhythmic agent as a promising lead compound for targeting the hEAG1 ion channel in cancer therapy: insights from molecular dynamics simulations. Chem Biol Drug Des 2016; 88:683-689. [PMID: 27254790 DOI: 10.1111/cbdd.12797] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/27/2016] [Accepted: 05/30/2016] [Indexed: 11/29/2022]
Abstract
Experimental evidence suggests that hERG and hEAG potassium channels may serve as important cancer therapy targets because either of the channel blockade or inactivation by different methods leads to inhibition of cancer cells growth and proliferation. However, there is no known hEAG specific blocker, and hERG blockade leads to adverse cardiac side effects, although it is currently used in treating certain types of arrhythmias. There have been some attempts to explain the channels blockade by clofilium, an antiarrhythmic agent, and the results lead to different possible binding modes. This study investigates for the first time the potential of using clofilium as a lead compound for finding a novel cancer therapy agent which may target ion channels. The implied findings from a comparative assessment of literature studies were verified using molecular dynamics simulations. The results indicate a particular structural difference between the two channels that could provide a novel and realistic way of using clofilium analogs which may target the hEAG1 ion channel in cancer therapy.
Collapse
Affiliation(s)
- Daniel Șterbuleac
- Department of Biology, Biophysics Laboratory, Alexandru Ioan Cuza University, Iași, Romania
| | - Călin Lucian Maniu
- Department of Biology, Biophysics Laboratory, Alexandru Ioan Cuza University, Iași, Romania.
| |
Collapse
|
224
|
Xu F, Wu X, Jiang LH, Zhao H, Pan J. An organelle K+ channel is required for osmoregulation in Chlamydomonas reinhardtii. J Cell Sci 2016; 129:3008-14. [PMID: 27311484 DOI: 10.1242/jcs.188441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/13/2016] [Indexed: 11/20/2022] Open
Abstract
Fresh water protozoa and algae face hypotonic challenges in their living environment. Many of them employ a contractile vacuole system to uptake excessive water from the cytoplasm and expel it to the environment to achieve cellular homeostasis. K(+), a major osmolyte in contractile vacuole, is predicted to create higher osmolarity for water influx. Molecular mechanisms for K(+) permeation through the plasma membrane have been well studied. However, how K(+) permeates organelles such as the contractile vacuole is not clear. Here, we show that the six-transmembrane K(+) channel KCN11 in Chlamydomonas is exclusively localized to contractile vacuole. Ectopic expression of KCN11 in HEK293T cells results in voltage-gated K(+) channel activity. Disruption of the gene or mutation of key residues for K(+) permeability of the channel leads to dysfunction of cell osmoregulation in very hypotonic conditions. The contractile cycle is inhibited in the mutant cells with a slower rate of contractile vacuole swelling, leading to cell death. These data demonstrate a new role for six-transmembrane K(+) channels in contractile vacuole functioning and provide further insights into osmoregulation mediated by the contractile vacuole.
Collapse
Affiliation(s)
- Feifei Xu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoan Wu
- Laboratory of Biomechanics, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Hucheng Zhao
- Laboratory of Biomechanics, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Junmin Pan
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong Province 266071, China
| |
Collapse
|
225
|
Cheng YY, Wright CM, Kirschner MB, Williams M, Sarun KH, Sytnyk V, Leshchynska I, Edelman JJ, Vallely MP, McCaughan BC, Klebe S, van Zandwijk N, Lin RCY, Reid G. KCa1.1, a calcium-activated potassium channel subunit alpha 1, is targeted by miR-17-5p and modulates cell migration in malignant pleural mesothelioma. Mol Cancer 2016; 15:44. [PMID: 27245839 PMCID: PMC4888473 DOI: 10.1186/s12943-016-0529-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/20/2016] [Indexed: 01/22/2023] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is an aggressive, locally invasive, cancer elicited by asbestos exposure and almost invariably a fatal diagnosis. To date, we are one of the leading laboratory that compared microRNA expression profiles in MPM and normal mesothelium samples in order to identify dysregulated microRNAs with functional roles in mesothelioma. We interrogated a significant collection of MPM tumors and normal pleural samples in our biobank in search for novel therapeutic targets. Methods Utilizing mRNA-microRNA correlations based on differential gene expression using Gene Set Enrichment Analysis (GSEA), we systematically combined publicly available gene expression datasets with our own MPM data in order to identify candidate targets for MPM therapy. Results We identified enrichment of target binding sites for the miR-17 and miR-30 families in both MPM tumors and cell lines. RT-qPCR revealed that members of both families were significantly downregulated in MPM tumors and cell lines. Interestingly, lower expression of miR-17-5p (P = 0.022) and miR-20a-5p (P = 0.026) was clearly associated with epithelioid histology. We interrogated the predicted targets of these differentially expressed microRNA families in MPM cell lines, and identified KCa1.1, a calcium-activated potassium channel subunit alpha 1 encoded by the KCNMA1 gene, as a target of miR-17-5p. KCa1.1 was overexpressed in MPM cells compared to the (normal) mesothelial line MeT-5A, and was also upregulated in patient tumor samples compared to normal mesothelium. Transfection of MPM cells with a miR-17-5p mimic or KCNMA1-specific siRNAs reduced mRNA expression of KCa1.1 and inhibited MPM cell migration. Similarly, treatment with paxilline, a small molecule inhibitor of KCa1.1, resulted in suppression of MPM cell migration. Conclusion These functional data implicating KCa1.1 in MPM cell migration support our integrative approach using MPM gene expression datasets to identify novel and potentially druggable targets. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0529-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuen Yee Cheng
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia
| | - Casey M Wright
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia
| | - Michaela B Kirschner
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia.,Division of Thoracic Surgery, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Marissa Williams
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia.,School of Medicine, University of Sydney, Sydney, NSW, 2006, Australia
| | - Kadir H Sarun
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Iryna Leshchynska
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - J James Edelman
- Cardiothoracic Surgical Unit, Royal Prince Alfred Hospital; The Baird Institute and Faculty of Medicine, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michael P Vallely
- Cardiothoracic Surgical Unit, Royal Prince Alfred Hospital; The Baird Institute and Faculty of Medicine, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Brian C McCaughan
- Sydney Cardiothoracic Surgeons, RPA Medical Centre, Sydney, NSW, 2050, Australia
| | - Sonja Klebe
- Department of Anatomical Pathology, Flinders Medical Centre, Adelaide, SA, 5042, Australia
| | - Nico van Zandwijk
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia.,School of Medicine, University of Sydney, Sydney, NSW, 2006, Australia
| | - Ruby C Y Lin
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia. .,School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Glen Reid
- Asbestos Diseases Research Institute, Gate 3, Hospital Road, Concord, Sydney, NSW, 2139, Australia. .,School of Medicine, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
226
|
Novel cell-free high-throughput screening method for pharmacological tools targeting K+ channels. Proc Natl Acad Sci U S A 2016; 113:5748-53. [PMID: 27091997 DOI: 10.1073/pnas.1602815113] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
K(+) channels, a superfamily of ∼80 members, control cell excitability, ion homeostasis, and many forms of cell signaling. Their malfunctions cause numerous diseases including neuronal disorders, cardiac arrhythmia, diabetes, and asthma. Here we present a novel liposome flux assay (LFA) that is applicable to most K(+) channels. It is robust, low cost, and high throughput. Using LFA, we performed small molecule screens on three different K(+) channels and identified new activators and inhibitors for biological research on channel function and for medicinal development. We further engineered a hERG (human ether-à-go-go-related gene) channel, which, when used in LFA, provides a highly sensitive (zero false negatives on 50 hERG-sensitive drugs) and highly specific (zero false positives on 50 hERG-insensitive drugs), low-cost hERG safety assay.
Collapse
|
227
|
Rivera-Torres IO, Jin TB, Cadene M, Chait BT, Poget SF. Discovery and characterisation of a novel toxin from Dendroaspis angusticeps, named Tx7335, that activates the potassium channel KcsA. Sci Rep 2016; 6:23904. [PMID: 27044983 PMCID: PMC4820689 DOI: 10.1038/srep23904] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
Abstract
Due to their central role in essential physiological processes, potassium channels are common targets for animal toxins. These toxins in turn are of great value as tools for studying channel function and as lead compounds for drug development. Here, we used a direct toxin pull-down assay with immobilised KcsA potassium channel to isolate a novel KcsA-binding toxin (called Tx7335) from eastern green mamba snake (Dendroaspis angusticeps) venom. Sequencing of the toxin by Edman degradation and mass spectrometry revealed a 63 amino acid residue peptide with 4 disulphide bonds that belongs to the three-finger toxin family, but with a unique modification of its disulphide-bridge scaffold. The toxin induces a dose-dependent increase in both open probabilities and mean open times on KcsA in artificial bilayers. Thus, it unexpectedly behaves as a channel activator rather than an inhibitor. A charybdotoxin-sensitive mutant of KcsA exhibits similar susceptibility to Tx7335 as wild-type, indicating that the binding site for Tx7335 is distinct from that of canonical pore-blocker toxins. Based on the extracellular location of the toxin binding site (far away from the intracellular pH gate), we propose that Tx7335 increases potassium flow through KcsA by allosterically reducing inactivation of the channel.
Collapse
Affiliation(s)
- Iván O. Rivera-Torres
- LaGuardia Community College, City University of New York, Long Island City, NY 11101, USA
| | - Tony B. Jin
- Department of Chemistry, CUNY Graduate Center and Institute for Macromolecular Assemblies, College of Staten Island, City University of New York, Staten Island, NY 10314, USA
| | | | | | - Sébastien F. Poget
- Department of Chemistry, CUNY Graduate Center and Institute for Macromolecular Assemblies, College of Staten Island, City University of New York, Staten Island, NY 10314, USA
| |
Collapse
|
228
|
Novoseletsky VN, Volyntseva AD, Shaitan KV, Kirpichnikov MP, Feofanov AV. Modeling of the Binding of Peptide Blockers to Voltage-Gated Potassium Channels: Approaches and Evidence. Acta Naturae 2016; 8:35-46. [PMID: 27437138 PMCID: PMC4947987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Indexed: 11/13/2022] Open
Abstract
Modeling of the structure of voltage-gated potassium (KV) channels bound to peptide blockers aims to identify the key amino acid residues dictating affinity and provide insights into the toxin-channel interface. Computational approaches open up possibilities for in silico rational design of selective blockers, new molecular tools to study the cellular distribution and functional roles of potassium channels. It is anticipated that optimized blockers will advance the development of drugs that reduce over activation of potassium channels and attenuate the associated malfunction. Starting with an overview of the recent advances in computational simulation strategies to predict the bound state orientations of peptide pore blockers relative to KV-channels, we go on to review algorithms for the analysis of intermolecular interactions, and then take a look at the results of their application.
Collapse
Affiliation(s)
- V. N. Novoseletsky
- M.V.Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bldg. 12, 119992 , Moscow, Russia
| | - A. D. Volyntseva
- M.V.Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bldg. 12, 119992 , Moscow, Russia
| | - K. V. Shaitan
- M.V.Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bldg. 12, 119992 , Moscow, Russia
| | - M. P. Kirpichnikov
- M.V.Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bldg. 12, 119992 , Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho- Maklaya str. 16/10, 117997, Moscow, Russia
| | - A. V. Feofanov
- M.V.Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bldg. 12, 119992 , Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho- Maklaya str. 16/10, 117997, Moscow, Russia
| |
Collapse
|
229
|
Durant F, Lobo D, Hammelman J, Levin M. Physiological controls of large-scale patterning in planarian regeneration: a molecular and computational perspective on growth and form. REGENERATION (OXFORD, ENGLAND) 2016; 3:78-102. [PMID: 27499881 PMCID: PMC4895326 DOI: 10.1002/reg2.54] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/18/2016] [Accepted: 02/22/2016] [Indexed: 12/12/2022]
Abstract
Planaria are complex metazoans that repair damage to their bodies and cease remodeling when a correct anatomy has been achieved. This model system offers a unique opportunity to understand how large-scale anatomical homeostasis emerges from the activities of individual cells. Much progress has been made on the molecular genetics of stem cell activity in planaria. However, recent data also indicate that the global pattern is regulated by physiological circuits composed of ionic and neurotransmitter signaling. Here, we overview the multi-scale problem of understanding pattern regulation in planaria, with specific focus on bioelectric signaling via ion channels and gap junctions (electrical synapses), and computational efforts to extract explanatory models from functional and molecular data on regeneration. We present a perspective that interprets results in this fascinating field using concepts from dynamical systems theory and computational neuroscience. Serving as a tractable nexus between genetic, physiological, and computational approaches to pattern regulation, planarian pattern homeostasis harbors many deep insights for regenerative medicine, evolutionary biology, and engineering.
Collapse
Affiliation(s)
- Fallon Durant
- Department of Biology, Allen Discovery Center at Tufts University, Tufts Center for Regenerative and Developmental BiologyTufts UniversityMA02155USA
| | - Daniel Lobo
- Department of Biological SciencesUniversity of MarylandBaltimore County, 1000 Hilltop CircleBaltimoreMD21250USA
| | - Jennifer Hammelman
- Department of Biology, Allen Discovery Center at Tufts University, Tufts Center for Regenerative and Developmental BiologyTufts UniversityMA02155USA
| | - Michael Levin
- Department of Biology, Allen Discovery Center at Tufts University, Tufts Center for Regenerative and Developmental BiologyTufts UniversityMA02155USA
| |
Collapse
|
230
|
Ohya S, Kanatsuka S, Hatano N, Kito H, Matsui A, Fujimoto M, Matsuba S, Niwa S, Zhan P, Suzuki T, Muraki K. Downregulation of the Ca(2+)-activated K(+) channel KC a3.1 by histone deacetylase inhibition in human breast cancer cells. Pharmacol Res Perspect 2016; 4:e00228. [PMID: 27069638 PMCID: PMC4804315 DOI: 10.1002/prp2.228] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/12/2016] [Indexed: 12/11/2022] Open
Abstract
The intermediate‐conductance Ca2+‐activated K+ channel KCa3.1 is involved in the promotion of tumor growth and metastasis, and is a potential therapeutic target and biomarker for cancer. Histone deacetylase inhibitors (HDACis) have considerable potential for cancer therapy, however, the effects of HDACis on ion channel expression have not yet been investigated in detail. The results of this study showed a significant decrease in KCa3.1 transcription by HDAC inhibition in the human breast cancer cell line YMB‐1, which functionally expresses KCa3.1. A treatment with the clinically available, class I, II, and IV HDAC inhibitor, vorinostat significantly downregulated KCa3.1 transcription in a concentration‐dependent manner, and the plasmalemmal expression of the KCa3.1 protein and its functional activity were correspondingly decreased. Pharmacological and siRNA‐based HDAC inhibition both revealed the involvement of HDAC2 and HDAC3 in KCa3.1 transcription through the same mechanism. The downregulation of KCa3.1 in YMB‐1 was not due to the upregulation of the repressor element‐1 silencing transcription factor, REST and the insulin‐like growth factor‐binding protein 5, IGFBP5. The significant decrease in KCa3.1 transcription by HDAC inhibition was also observed in the KCa3.1‐expressing human prostate cancer cell line, PC‐3. These results suggest that vorinostat and the selective HDACis for HDAC2 and/or HDAC3 are effective drug candidates for KCa3.1‐overexpressing cancers.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Saki Kanatsuka
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology School of Pharmacy Aichi-Gakuin University Nagoya 464-8650 Japan
| | - Hiroaki Kito
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Azusa Matsui
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Mayu Fujimoto
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Sayo Matsuba
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Satomi Niwa
- Department of Pharmacology Division of Pathological Sciences Kyoto Pharmaceutical University Kyoto 607-8414 Japan
| | - Peng Zhan
- Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto 606-0823 Japan
| | - Takayoshi Suzuki
- Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto 606-0823 Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology School of Pharmacy Aichi-Gakuin University Nagoya 464-8650 Japan
| |
Collapse
|
231
|
Fernández-Valle Á, Rodrigo JP, García-Pedrero JM, Rodríguez-Santamarta T, Allonca E, Lequerica-Fernández P, de Vicente JC. Expression of the voltage-gated potassium channel Kv3.4 in oral leucoplakias and oral squamous cell carcinomas. Histopathology 2016; 69:91-8. [DOI: 10.1111/his.12917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/05/2015] [Indexed: 11/27/2022]
Affiliation(s)
- Álvaro Fernández-Valle
- Department of Oral and Maxillofacial Surgery; Hospital Universitario Central de Asturias (HUCA); Oviedo Asturias Spain
| | - Juan Pablo Rodrigo
- Department of Otolaryngology; Hospital Universitario Central de Asturias (HUCA); Oviedo Asturias Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA); University of Oviedo; Oviedo Asturias Spain
| | - Juana M García-Pedrero
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA); University of Oviedo; Oviedo Asturias Spain
| | - Tania Rodríguez-Santamarta
- Department of Oral and Maxillofacial Surgery; Hospital Universitario Central de Asturias (HUCA); Oviedo Asturias Spain
| | - Eva Allonca
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA); University of Oviedo; Oviedo Asturias Spain
| | | | - Juan Carlos de Vicente
- Department of Oral and Maxillofacial Surgery; Hospital Universitario Central de Asturias (HUCA); Oviedo Asturias Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA); University of Oviedo; Oviedo Asturias Spain
| |
Collapse
|
232
|
Ohya S, Kito H, Hatano N, Muraki K. Recent advances in therapeutic strategies that focus on the regulation of ion channel expression. Pharmacol Ther 2016; 160:11-43. [PMID: 26896566 DOI: 10.1016/j.pharmthera.2016.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of different ion channel types are involved in cell signaling networks, and homeostatic regulatory mechanisms contribute to the control of ion channel expression. Profiling of global gene expression using microarray technology has recently provided novel insights into the molecular mechanisms underlying the homeostatic and pathological control of ion channel expression. It has demonstrated that the dysregulation of ion channel expression is associated with the pathogenesis of neural, cardiovascular, and immune diseases as well as cancers. In addition to the transcriptional, translational, and post-translational regulation of ion channels, potentially important evidence on the mechanisms controlling ion channel expression has recently been accumulated. The regulation of alternative pre-mRNA splicing is therefore a novel therapeutic strategy for the treatment of dominant-negative splicing disorders. Epigenetic modification plays a key role in various pathological conditions through the regulation of pluripotency genes. Inhibitors of pre-mRNA splicing and histone deacetyalase/methyltransferase have potential as potent therapeutic drugs for cancers and autoimmune and inflammatory diseases. Moreover, membrane-anchoring proteins, lysosomal and proteasomal degradation-related molecules, auxiliary subunits, and pharmacological agents alter the protein folding, membrane trafficking, and post-translational modifications of ion channels, and are linked to expression-defect channelopathies. In this review, we focused on recent insights into the transcriptional, spliceosomal, epigenetic, and proteasomal regulation of ion channel expression: Ca(2+) channels (TRPC/TRPV/TRPM/TRPA/Orai), K(+) channels (voltage-gated, KV/Ca(2+)-activated, KCa/two-pore domain, K2P/inward-rectifier, Kir), and Ca(2+)-activated Cl(-) channels (TMEM16A/TMEM16B). Furthermore, this review highlights expression of these ion channels in expression-defect channelopathies.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan.
| |
Collapse
|
233
|
Cervera J, Alcaraz A, Mafe S. Bioelectrical Signals and Ion Channels in the Modeling of Multicellular Patterns and Cancer Biophysics. Sci Rep 2016; 6:20403. [PMID: 26841954 PMCID: PMC4740742 DOI: 10.1038/srep20403] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023] Open
Abstract
Bioelectrical signals and ion channels are central to spatial patterns in cell ensembles, a problem of fundamental interest in positional information and cancer processes. We propose a model for electrically connected cells based on simple biological concepts: i) the membrane potential of a single cell characterizes its electrical state; ii) the long-range electrical coupling of the multicellular ensemble is realized by a network of gap junction channels between neighboring cells; and iii) the spatial distribution of an external biochemical agent can modify the conductances of the ion channels in a cell membrane and the multicellular electrical state. We focus on electrical effects in small multicellular ensembles, ignoring slow diffusional processes. The spatio-temporal patterns obtained for the local map of cell electric potentials illustrate the normalization of regions with abnormal cell electrical states. The effects of intercellular coupling and blocking of specific channels on the electrical patterns are described. These patterns can regulate the electrically-induced redistribution of charged nanoparticles over small regions of a model tissue. The inclusion of bioelectrical signals provides new insights for the modeling of cancer biophysics because collective multicellular states show electrical coupling mechanisms that are not readily deduced from biochemical descriptions at the individual cell level.
Collapse
Affiliation(s)
- Javier Cervera
- Dept. de Termodinàmica, Facultat de Física, Universitat de València, E-46100 Burjassot, Spain
| | - Antonio Alcaraz
- Dept. de Física, Laboratori de Biofísica Molecular, Universitat “Jaume I”, E-12080 Castelló, Spain
| | - Salvador Mafe
- Dept. de Termodinàmica, Facultat de Física, Universitat de València, E-46100 Burjassot, Spain
| |
Collapse
|
234
|
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
|
235
|
Crottès D, Rapetti-Mauss R, Alcaraz-Perez F, Tichet M, Gariano G, Martial S, Guizouarn H, Pellissier B, Loubat A, Popa A, Paquet A, Presta M, Tartare-Deckert S, Cayuela ML, Martin P, Borgese F, Soriani O. SIGMAR1 Regulates Membrane Electrical Activity in Response to Extracellular Matrix Stimulation to Drive Cancer Cell Invasiveness. Cancer Res 2016; 76:607-18. [PMID: 26645564 DOI: 10.1158/0008-5472.can-15-1465] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/27/2015] [Indexed: 01/12/2023]
Abstract
The sigma 1 receptor (Sig1R) is a stress-activated chaperone that regulates ion channels and is associated with pathologic conditions, such as stroke, neurodegenerative diseases, and addiction. Aberrant expression levels of ion channels and Sig1R have been detected in tumors and cancer cells, such as myeloid leukemia and colorectal cancer, but the link between ion channel regulation and Sig1R overexpression during malignancy has not been established. In this study, we found that Sig1R dynamically controls the membrane expression of the human voltage-dependent K(+) channel human ether-à-go-go-related gene (hERG) in myeloid leukemia and colorectal cancer cell lines. Sig1R promoted the formation of hERG/β1-integrin signaling complexes upon extracellular matrix stimulation, triggering the activation of the PI3K/AKT pathway. Consequently, the presence of Sig1R in cancer cells increased motility and VEGF secretion. In vivo, Sig1R expression enhanced the aggressiveness of tumor cells by potentiating invasion and angiogenesis, leading to poor survival. Collectively, our findings highlight a novel function for Sig1R in mediating cross-talk between cancer cells and their microenvironment, thus driving oncogenesis by shaping cellular electrical activity in response to extracellular signals. Given the involvement of ion channels in promoting several hallmarks of cancer, our study also offers a potential strategy to therapeutically target ion channel function through Sig1R inhibition.
Collapse
Affiliation(s)
- David Crottès
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France. Department of Physiology, University of California, San Francisco, San Francisco, California
| | - Raphael Rapetti-Mauss
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Francisca Alcaraz-Perez
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Mélanie Tichet
- Université Nice Sophia Antipolis, C3M, Inserm U1065, Nice, France
| | - Giuseppina Gariano
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sonia Martial
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Hélène Guizouarn
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Bernard Pellissier
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Agnès Loubat
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Alexandra Popa
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Agnès Paquet
- Université Nice Sophia Antipolis, IPMC, CNRS UMR7275, Sophia Antipolis, France
| | - Marco Presta
- Unit of Oncology and Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Maria Luisa Cayuela
- Telomerase, Aging and Cancer Group, Research Unit, Department of Surgery, CIBERehd, University Hospital "Virgen de la Arrixaca", Murcia, Spain. Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Patrick Martin
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Franck Borgese
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France
| | - Olivier Soriani
- Université Nice Sophia Antipolis, iBV, Nice, France. CNRS, iBV, UMR7277, Nice, France. INSERM U1091, Nice, France.
| |
Collapse
|
236
|
Lakomá J, Donadio V, Liguori R, Caprini M. Characterization of Human Dermal Fibroblasts in Fabry Disease. J Cell Physiol 2016; 231:192-203. [PMID: 26058984 DOI: 10.1002/jcp.25072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/05/2015] [Indexed: 12/14/2022]
Abstract
Fabry disease (FD) is a hereditary X-linked metabolic lysosomal storage disorder due to insufficient amounts or a complete lack of the lysosomal enzyme α-galactosidase A (α-GalA). The loss of α-GalA activity leads to an abnormal accumulation of globotriaosylcerami (Gb3) in lysosomes and other cellular components of different tissues and cell types, affecting the cell function. However, whether these biochemical alterations also modify functional processes associated to the cell mitotic ability is still unknown. The goal of the present study was to characterize lineages of human dermal fibroblasts (HDFs) of FD patients and healthy controls focusing on Gb3 accumulation, expression of chloride channels that regulate proliferation, and proliferative activity. The biochemical and functional analyses indicate the existence of quantitative differences in some but not all the parameters of cytoskeletal organization, proliferation, and differentiation processes.
Collapse
Affiliation(s)
- Jarmila Lakomá
- Laboratory of Human General Physiology, Department of Pharmacy Biotechnology FaBiT, University of Bologna, Bologna, Italy
| | - Vincenzo Donadio
- IRCCS Institute of Neurological Sciences, AUSL Bologna, Bologna, Italy
| | - Rocco Liguori
- IRCCS Institute of Neurological Sciences, AUSL Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Marco Caprini
- Laboratory of Human General Physiology, Department of Pharmacy Biotechnology FaBiT, University of Bologna, Bologna, Italy
| |
Collapse
|
237
|
Kaczorowski G, Garcia M. Developing Molecular Pharmacology of BK Channels for Therapeutic Benefit. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:439-75. [DOI: 10.1016/bs.irn.2016.02.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
238
|
Yin MZ, Park SW, Kang TW, Kim KS, Yoo HY, Lee J, Hah JH, Sung MH, Kim SJ. Activation of K(+) channel by 1-EBIO rescues the head and neck squamous cell carcinoma cells from Ca(2+) ionophore-induced cell death. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 20:25-33. [PMID: 26807020 PMCID: PMC4722188 DOI: 10.4196/kjpp.2016.20.1.25] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/14/2015] [Accepted: 08/14/2015] [Indexed: 12/11/2022]
Abstract
Ion channels in carcinoma and their roles in cell proliferation are drawing attention. Intracellular Ca2+ ([Ca2+]i)-dependent signaling affects the fate of cancer cells. Here we investigate the role of Ca2+-activated K+ channel (SK4) in head and neck squamous cell carcinoma cells (HNSCCs) of different cell lines; SNU-1076, OSC-19 and HN5. Treatment with 1 µM ionomycin induced cell death in all the three cell lines. Whole-cell patch clamp study suggested common expressions of Ca2+-activated Cl- channels (Ano-1) and Ca2+-activated nonselective cation channels (CAN). 1-EBIO, an activator of SK4, induced outward K+ current (ISK4) in SNU-1076 and OSC-19. In HN5, ISK4 was not observed or negligible. The 1-EBIO-induced current was abolished by TRAM-34, a selective SK4 blocker. Interestingly, the ionomycin-induced cell death was effectively prevented by 1-EBIO in SNU-1076 and OSC-19, and the rescue effect was annihilated by combined TRAM-34. Consistent with the lower level of ISK4, the rescue by 1-EBIO was least effective in HN5. The results newly demonstrate the role of SK4 in the fate of HNSCCs under the Ca2+ overloaded condition. Pharmacological modulation of SK4 might provide an intriguing novel tool for the anti-cancer strategy in HNSCC.
Collapse
Affiliation(s)
- Ming Zhe Yin
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seok-Woo Park
- Department of Otolaryngology, Seoul National University Hospital, Seoul 03080, Korea.; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Tae Wook Kang
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Kyung Soo Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hae Young Yoo
- Chung-Ang University Red Cross College of Nursing, Seoul 06974, Korea
| | - Junho Lee
- Department of Otolaryngology, Seoul National University Hospital, Seoul 03080, Korea
| | - J Hun Hah
- Department of Otolaryngology, Seoul National University Hospital, Seoul 03080, Korea.; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Myung Hun Sung
- Department of Otolaryngology, Seoul National University Hospital, Seoul 03080, Korea.; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
239
|
Delgado-Ramírez M, Morán-Zendejas R, Aréchiga-Figueroa IA, Toro-Castillo C, Ramírez-Martínez JF, Rodríguez-Menchaca AA. Modulation of the voltage-gated potassium channel Kv2.1 by the anti-tumor alkylphospholipid perifosine. Pharmacol Rep 2015; 68:457-61. [PMID: 26922553 DOI: 10.1016/j.pharep.2015.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/10/2015] [Accepted: 11/13/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND The aim of the present study was to assess the effects of perifosine-a third generation alkylphospholipid analog with anti-tumor properties-on the activity of Kv2.1 channels. METHODS The whole-cell patch clamp technique was applied to follow the modulatory effect of perifosine on Kv2.1 channels expressed in HEK293 cells. RESULTS Obtained data provide evidence that perifosine application decreases the whole cell Kv2.1 currents in a concentration-independent manner. Perifosine induces a hyperpolarizing shift in the voltage dependence of Kv2.1 channels inactivation without altering the voltage dependence of channels activation. The kinetics of Kv2.1 closed-state inactivation was accelerated by perifosine, with no significant effects on the recovery rate from inactivation. CONCLUSIONS Taken together, these results show that perifosine modified the Kv2.1 inactivation gating resulting in a decrease of the current amplitude. These data will help to elucidate the mechanism of action of this promising anti-cancer drug on ion channels and their possible implications.
Collapse
Affiliation(s)
- Mayra Delgado-Ramírez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Rita Morán-Zendejas
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Ivan A Aréchiga-Figueroa
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Carmen Toro-Castillo
- Departamento de Ciencias Computacionales, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Jalisco, México
| | - Juan F Ramírez-Martínez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Aldo A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| |
Collapse
|
240
|
Cang C, Aranda K, Seo YJ, Gasnier B, Ren D. TMEM175 Is an Organelle K(+) Channel Regulating Lysosomal Function. Cell 2015; 162:1101-12. [PMID: 26317472 DOI: 10.1016/j.cell.2015.08.002] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/10/2015] [Accepted: 07/13/2015] [Indexed: 12/15/2022]
Abstract
Potassium is the most abundant ion to face both plasma and organelle membranes. Extensive research over the past seven decades has characterized how K(+) permeates the plasma membrane to control fundamental processes such as secretion, neuronal communication, and heartbeat. However, how K(+) permeates organelles such as lysosomes and endosomes is unknown. Here, we directly recorded organelle K(+) conductance and discovered a major K(+)-selective channel KEL on endosomes and lysosomes. KEL is formed by TMEM175, a protein with unknown function. Unlike any of the ∼80 plasma membrane K(+) channels, TMEM175 has two repeats of 6-transmembrane-spanning segments and has no GYG K(+) channel sequence signature-containing, pore-forming P loop. Lysosomes lacking TMEM175 exhibit no K(+) conductance, have a markedly depolarized ΔΨ and little sensitivity to changes in [K(+)], and have compromised luminal pH stability and abnormal fusion with autophagosomes during autophagy. Thus, TMEM175 comprises a K(+) channel that underlies the molecular mechanism of lysosomal K(+) permeability.
Collapse
Affiliation(s)
- Chunlei Cang
- Department of Biology, University of Pennsylvania, 415 South University Avenue, Philadelphia, PA 19104, USA
| | - Kimberly Aranda
- Department of Biology, University of Pennsylvania, 415 South University Avenue, Philadelphia, PA 19104, USA
| | - Young-jun Seo
- Department of Biology, University of Pennsylvania, 415 South University Avenue, Philadelphia, PA 19104, USA
| | - Bruno Gasnier
- Paris Descartes University, Sorbonne Paris Cité, Neurophotonics Laboratory, Centre National de la Recherche Scientifique UMR8250, 45 rue des Saints Pères, 75006 Paris, France
| | - Dejian Ren
- Department of Biology, University of Pennsylvania, 415 South University Avenue, Philadelphia, PA 19104, USA.
| |
Collapse
|
241
|
Ryland KE, Hawkins AG, Weisenberger DJ, Punj V, Borinstein SC, Laird PW, Martens JR, Lawlor ER. Promoter Methylation Analysis Reveals That KCNA5 Ion Channel Silencing Supports Ewing Sarcoma Cell Proliferation. Mol Cancer Res 2015; 14:26-34. [PMID: 26573141 DOI: 10.1158/1541-7786.mcr-15-0343] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/04/2015] [Indexed: 02/07/2023]
Abstract
UNLABELLED Polycomb proteins are essential regulators of gene expression in stem cells and development. They function to reversibly repress gene transcription via posttranslational modification of histones and chromatin compaction. In many human cancers, genes that are repressed by polycomb in stem cells are subject to more stable silencing via DNA methylation of promoter CpG islands. Ewing sarcoma is an aggressive bone and soft-tissue tumor that is characterized by overexpression of polycomb proteins. This study investigates the DNA methylation status of polycomb target gene promoters in Ewing sarcoma tumors and cell lines and observes that the promoters of differentiation genes are frequent targets of CpG-island DNA methylation. In addition, the promoters of ion channel genes are highly differentially methylated in Ewing sarcoma compared with nonmalignant adult tissues. Ion channels regulate a variety of biologic processes, including proliferation, and dysfunction of these channels contributes to tumor pathogenesis. In particular, reduced expression of the voltage-gated Kv1.5 channel has been implicated in tumor progression. These data show that DNA methylation of the KCNA5 promoter contributes to stable epigenetic silencing of the Kv1.5 channel. This epigenetic repression is reversed by exposure to the DNA methylation inhibitor decitabine, which inhibits Ewing sarcoma cell proliferation through mechanisms that include restoration of the Kv1.5 channel function. IMPLICATIONS This study demonstrates that promoters of ion channels are aberrantly methylated in Ewing sarcoma and that epigenetic silencing of KCNA5 contributes to tumor cell proliferation, thus providing further evidence of the importance of ion channel dysregulation to tumorigenesis.
Collapse
Affiliation(s)
- Katherine E Ryland
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan. Translational Oncology Program, University of Michigan, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Allegra G Hawkins
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California. Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Vasu Punj
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Peter W Laird
- Van Andel Research Institute, Grand Rapids, Michigan
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida
| | - Elizabeth R Lawlor
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan, Ann Arbor, Michigan. Department of Pathology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
242
|
Cobb MM, Austin DC, Sack JT, Trimmer JS. Cell Cycle-dependent Changes in Localization and Phosphorylation of the Plasma Membrane Kv2.1 K+ Channel Impact Endoplasmic Reticulum Membrane Contact Sites in COS-1 Cells. J Biol Chem 2015; 290:29189-201. [PMID: 26442584 DOI: 10.1074/jbc.m115.690198] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Indexed: 12/22/2022] Open
Abstract
The plasma membrane (PM) comprises distinct subcellular domains with diverse functions that need to be dynamically coordinated with intracellular events, one of the most impactful being mitosis. The Kv2.1 voltage-gated potassium channel is conditionally localized to large PM clusters that represent specialized PM:endoplasmic reticulum membrane contact sites (PM:ER MCS), and overexpression of Kv2.1 induces more exuberant PM:ER MCS in neurons and in certain heterologous cell types. Localization of Kv2.1 at these contact sites is dynamically regulated by changes in phosphorylation at one or more sites located on its large cytoplasmic C terminus. Here, we show that Kv2.1 expressed in COS-1 cells undergoes dramatic cell cycle-dependent changes in its PM localization, having diffuse localization in interphase cells, and robust clustering during M phase. The mitosis-specific clusters of Kv2.1 are localized to PM:ER MCS, and M phase clustering of Kv2.1 induces more extensive PM:ER MCS. These cell cycle-dependent changes in Kv2.1 localization and the induction of PM:ER MCS are accompanied by increased mitotic Kv2.1 phosphorylation at several C-terminal phosphorylation sites. Phosphorylation of exogenously expressed Kv2.1 is significantly increased upon metaphase arrest in COS-1 and CHO cells, and in a pancreatic β cell line that express endogenous Kv2.1. The M phase clustering of Kv2.1 at PM:ER MCS in COS-1 cells requires the same C-terminal targeting motif needed for conditional Kv2.1 clustering in neurons. The cell cycle-dependent changes in localization and phosphorylation of Kv2.1 were not accompanied by changes in the electrophysiological properties of Kv2.1 expressed in CHO cells. Together, these results provide novel insights into the cell cycle-dependent changes in PM protein localization and phosphorylation.
Collapse
Affiliation(s)
- Melanie M Cobb
- From the Departments of Neurobiology, Physiology, and Behavior
| | | | - Jon T Sack
- Physiology and Membrane Biology, and Anesthesiology and Pain Medicine, University of California Davis School of Medicine, Davis, California 95616
| | - James S Trimmer
- From the Departments of Neurobiology, Physiology, and Behavior, Physiology and Membrane Biology, and
| |
Collapse
|
243
|
Kong X, Su F, Zhang L, Yaron J, Lee F, Shi Z, Tian Y, Meldrum DR. A highly selective mitochondria-targeting fluorescent K(+) sensor. Angew Chem Int Ed Engl 2015; 54:12053-7. [PMID: 26302172 PMCID: PMC4815426 DOI: 10.1002/anie.201506038] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Indexed: 11/08/2022]
Abstract
Regulation of intracellular potassium (K(+) ) concentration plays a key role in metabolic processes. So far, only a few intracellular K(+) sensors have been developed. The highly selective fluorescent K(+) sensor KS6 for monitoring K(+) ion dynamics in mitochondria was produced by coupling triphenylphosphonium, borondipyrromethene (BODIPY), and triazacryptand (TAC). KS6 shows a good response to K(+) in the range 30-500 mM, a large dynamic range (Fmax /F0 ≈130), high brightness (ϕf =14.4 % at 150 mM of K(+) ), and insensitivity to both pH in the range 5.5-9.0 and other metal ions under physiological conditions. Colocalization tests of KS6 with MitoTracker Green confirmed its predominant localization in the mitochondria of HeLa and U87MG cells. K(+) efflux/influx in the mitochondria was observed upon stimulation with ionophores, nigericin, or ionomycin. KS6 is thus a highly selective semiquantitative K(+) sensor suitable for the study of mitochondrial potassium flux in live cells.
Collapse
Affiliation(s)
- Xiangxing Kong
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA)
| | - Fengyu Su
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA)
| | - Liqiang Zhang
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA)
| | - Jordan Yaron
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA)
| | - Fred Lee
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA)
| | - Zhengwei Shi
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA)
| | - Yanqing Tian
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA).
- Department of Materials Science and Engineering, South University of Science and Technology of China, No. 1088, Xueyuan Rd., Xili, Nanshan District, Shenzhen, Guangdong, 518055 (China).
| | - Deirdre R Meldrum
- Center for Biosignatures Discovery Automation, Biodesign Institute, Arizona State University, 1001 S. McAlister Ave., P.O. Box 876501, Tempe, AZ 85287 (USA).
| |
Collapse
|
244
|
Villanueva S, Burgos J, López-Cayuqueo KI, Lai KMV, Valenzuela DM, Cid LP, Sepúlveda FV. Cleft Palate, Moderate Lung Developmental Retardation and Early Postnatal Lethality in Mice Deficient in the Kir7.1 Inwardly Rectifying K+ Channel. PLoS One 2015; 10:e0139284. [PMID: 26402555 PMCID: PMC4581704 DOI: 10.1371/journal.pone.0139284] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/09/2015] [Indexed: 12/15/2022] Open
Abstract
Kir7.1 is an inwardly rectifying K+ channel of the Kir superfamily encoded by the kcnj13 gene. Kir7.1 is present in epithelial tissues where it colocalizes with the Na+/K+-pump probably serving to recycle K+ taken up by the pump. Human mutations affecting Kir7.1 are associated with retinal degeneration diseases. We generated a mouse lacking Kir7.1 by ablation of the Kcnj13 gene. Homozygous mutant null mice die hours after birth and show cleft palate and moderate retardation in lung development. Kir7.1 is expressed in the epithelium covering the palatal processes at the time at which palate sealing takes place and our results suggest it might play an essential role in late palatogenesis. Our work also reveals a second unexpected role in the development and the physiology of the respiratory system, where Kir7.1 is expressed in epithelial cells all along the respiratory tree.
Collapse
Affiliation(s)
| | - Johanna Burgos
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Doctorado en Ciencias Veterinarias de la Universidad Austral de Chile, Valdivia, Chile
| | | | - Ka-Man Venus Lai
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - David M. Valenzuela
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - L. Pablo Cid
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | |
Collapse
|
245
|
Beyenbach KW, Yu Y, Piermarini PM, Denton J. Targeting renal epithelial channels for the control of insect vectors. Tissue Barriers 2015; 3:e1081861. [PMID: 26716074 DOI: 10.1080/21688370.2015.1081861] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/04/2015] [Accepted: 08/07/2015] [Indexed: 12/14/2022] Open
Abstract
Three small molecules were identified in high throughput screens that 1) block renal inward rectifier potassium (Kir) channels of Aedes aegypti expressed in HEK cells and Xenopus oocytes, 2) inhibit the secretion of KCl but not NaCl in isolated Malpighian tubules, and after injection into the hemolymph, 3) inhibit KCl excretion in vivo, and 4) render mosquitoes flightless or dead within 24h. Some mosquitoes had swollen abdomens at death consistent with renal failure. VU625, the most potent and promising small molecule for development as mosquitocide, inhibits AeKir1-mediated currents with an IC50 less than 100 nM. It is highly selective for AeKir1 over mammalian Kir channels, and it affects only 3 of 68 mammalian membrane proteins. These results document 1) renal failure as a new mode-of-action for mosquitocide development, 2) renal Kir channels as molecular target for inducing renal failure, and 3) the promise of the discovery and development of new species-specific insecticides.
Collapse
Affiliation(s)
- Klaus W Beyenbach
- Department of Biomedical Sciences; Cornell University ; Ithaca, NY USA
| | - Yasong Yu
- College of Medicine; SUNY Downstate Medical Center ; Brooklyn, NY USA
| | - Peter M Piermarini
- Department of Entomology; Ohio Agricultural Research and Development Center; The Ohio State University ; Wooster, OH USA
| | - Jerod Denton
- Department of Anesthesiology; Vanderbilt University School of Medicine ; Nashville, TN USA
| |
Collapse
|
246
|
Kong X, Su F, Zhang L, Yaron J, Lee F, Shi Z, Tian Y, Meldrum DR. A Highly Selective Mitochondria-Targeting Fluorescent K+Sensor. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201506038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
247
|
Hermann A, Sitdikova GF, Weiger TM. Oxidative Stress and Maxi Calcium-Activated Potassium (BK) Channels. Biomolecules 2015; 5:1870-911. [PMID: 26287261 PMCID: PMC4598779 DOI: 10.3390/biom5031870] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/13/2023] Open
Abstract
All cells contain ion channels in their outer (plasma) and inner (organelle) membranes. Ion channels, similar to other proteins, are targets of oxidative impact, which modulates ion fluxes across membranes. Subsequently, these ion currents affect electrical excitability, such as action potential discharge (in neurons, muscle, and receptor cells), alteration of the membrane resting potential, synaptic transmission, hormone secretion, muscle contraction or coordination of the cell cycle. In this chapter we summarize effects of oxidative stress and redox mechanisms on some ion channels, in particular on maxi calcium-activated potassium (BK) channels which play an outstanding role in a plethora of physiological and pathophysiological functions in almost all cells and tissues. We first elaborate on some general features of ion channel structure and function and then summarize effects of oxidative alterations of ion channels and their functional consequences.
Collapse
Affiliation(s)
- Anton Hermann
- Department of Cell Biology, Division of Cellular and Molecular Neurobiology, University of Salzburg, Salzburg 5020, Austria.
| | - Guzel F Sitdikova
- Department of Physiology of Man and Animals, Kazan Federal University, Kazan 420008, Russia.
| | - Thomas M Weiger
- Department of Cell Biology, Division of Cellular and Molecular Neurobiology, University of Salzburg, Salzburg 5020, Austria.
| |
Collapse
|
248
|
EAG2 potassium channel with evolutionarily conserved function as a brain tumor target. Nat Neurosci 2015; 18:1236-46. [PMID: 26258683 DOI: 10.1038/nn.4088] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/15/2015] [Indexed: 12/15/2022]
Abstract
Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.
Collapse
|
249
|
Rao VR, Perez-Neut M, Kaja S, Gentile S. Voltage-gated ion channels in cancer cell proliferation. Cancers (Basel) 2015; 7:849-75. [PMID: 26010603 PMCID: PMC4491688 DOI: 10.3390/cancers7020813] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/12/2015] [Indexed: 12/22/2022] Open
Abstract
Changes of the electrical charges across the surface cell membrane are absolutely necessary to maintain cellular homeostasis in physiological as well as in pathological conditions. The opening of ion channels alter the charge distribution across the surface membrane as they allow the diffusion of ions such as K+, Ca++, Cl.
Collapse
Affiliation(s)
- Vidhya R Rao
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago 2160 S. 1s tAve, Maywood, IL 60153, USA.
| | - Mathew Perez-Neut
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago 2160 S. 1s tAve, Maywood, IL 60153, USA.
| | - Simon Kaja
- Department of Ophthalmology and Vision Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes St., Kansas City, MO 64108, USA.
| | - Saverio Gentile
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago 2160 S. 1s tAve, Maywood, IL 60153, USA.
| |
Collapse
|
250
|
Kale VP, Amin SG, Pandey MK. Targeting ion channels for cancer therapy by repurposing the approved drugs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2747-55. [PMID: 25843679 DOI: 10.1016/j.bbamem.2015.03.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 03/18/2015] [Accepted: 03/27/2015] [Indexed: 12/21/2022]
Abstract
Ion channels have been shown to be involved in oncogenesis and efforts are being poured in to target the ion channels. There are many clinically approved drugs with ion channels as "off" targets. The question is, can these drugs be repurposed to inhibit ion channels for cancer treatment? Repurposing of drugs will not only save investors' money but also result in safer drugs for cancer patients. Advanced bioinformatics techniques and availability of a plethora of open access data on FDA approved drugs for various indications and omics data of large number of cancer types give a ray of hope to look for possibility of repurposing those drugs for cancer treatment. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Vijay Pralhad Kale
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Shantu G Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Manoj K Pandey
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|