1
|
Thi Hong Van N, Hyun Nam J. Intermediate conductance calcium-activated potassium channel (KCa3.1) in cancer: Emerging roles and therapeutic potentials. Biochem Pharmacol 2024; 230:116573. [PMID: 39396649 DOI: 10.1016/j.bcp.2024.116573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The KCa3.1 channel (also known as the KCNN4, IK1, or SK4 channel) is an intermediate-conductance calcium-activated potassium channel that regulates the membrane potential and maintains calcium homeostasis. Recently, KCa3.1 channels have attracted increasing attention because of their diverse roles in various types of cancers. In cancer cells, KCa3.1 channels regulate key processes, including cell proliferation, cell cycle, migration, invasion, tumor microenvironments, and therapy resistance. In addition, abnormal KCa3.1 expression in cancers is utilized to distinguish between tumor and normal tissues, classify cancer stages, and predict patient survival outcomes. This review comprehensively examines the current understanding of the contribution of KCa3.1 channels to tumor formation, metastasis, and its mechanisms. We evaluated the potential of KCa3.1 as a biomarker for cancer diagnosis and prognosis. Finally, we discuss the advances and challenges of applying KCa3.1 modulators in cancer treatment and propose approaches to overcome these obstacles. In summary, this review highlights the importance of this ion channel as a potent therapeutic target and prognostic biomarker of cancer.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea.
| |
Collapse
|
2
|
Hapeman JD, Galwa R, Carneiro CS, Nedelcu AM. In vitro evidence for the potential of EGFR inhibitors to decrease the TGF-β1-induced dispersal of circulating tumour cell clusters mediated by EGFR overexpression. Sci Rep 2024; 14:19980. [PMID: 39198539 PMCID: PMC11358385 DOI: 10.1038/s41598-024-70358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Most cancer-related deaths are due to the spread of tumour cells throughout the body-a process known as metastasis. While in the vasculature, these cells are referred to as circulating tumour cells (CTCs) and can be found as either single cells or clusters of cells (often including platelets), with the latter having the highest metastatic potential. However, the biology of CTC clusters is poorly understood, and there are no therapies that specifically target them. We previously developed an in vitro model system for CTC clusters and proposed a new extravasation model that involves cluster dissociation, adherence, and single-cell invasion in response to TGF-β1 released by platelets. Here, we investigated TGF-β1-induced gene expression changes in this model, focusing on genes for which targeted drugs are available. In addition to the upregulation of the TGF-β1 signalling pathway, we found that (i) genes in the EGF/EGFR pathway, including those coding for EGFR and several EGFR ligands, were also induced, and (ii) Erlotinib and Osimertinib, two therapeutic EGFR/tyrosine kinase inhibitors, decreased the TGF-β1-induced adherence and invasion of the CTC cluster-like line despite the line expressing wild-type EGFR. Overall, we suggest that EGFR inhibitors have the potential to decrease the dispersal of CTC clusters that respond to TGF-β1 and overexpress EGFR (irrespective of its status) and thus could improve patient survival.
Collapse
Affiliation(s)
- Jorian D Hapeman
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Rakshit Galwa
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Caroline S Carneiro
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Aurora M Nedelcu
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada.
| |
Collapse
|
3
|
Zhang Y, Yan H, Wei Y, Wei X. Decoding mitochondria's role in immunity and cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189107. [PMID: 38734035 DOI: 10.1016/j.bbcan.2024.189107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The functions of mitochondria, including energy production and biomolecule synthesis, have been known for a long time. Given the rising incidence of cancer, the role of mitochondria in cancer has become increasingly popular. Activated by components released by mitochondria, various pathways interact with each other to induce immune responses to protect organisms from attack. However, mitochondria play dual roles in the progression of cancer. Abnormalities in proteins, which are the elementary structures of mitochondria, are closely linked with oncogenesis. Both the aberrant accumulation of intermediates and mutations in enzymes result in the generation and progression of cancer. Therefore, targeting mitochondria to treat cancer may be a new strategy. Several drugs aimed at inhibiting mutated enzymes and accumulated intermediates have been tested clinically. Here, we discuss the current understanding of mitochondria in cancer and the interactions between mitochondrial functions, immune responses, and oncogenesis. Furthermore, we discuss mitochondria as hopeful targets for cancer therapy, providing insights into the progression of future therapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Hong Yan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| |
Collapse
|
4
|
Zhang L, Ren C, Liu J, Huang S, Wu C, Zhang J. Development and therapeutic implications of small molecular inhibitors that target calcium-related channels in tumor treatment. Drug Discov Today 2024; 29:103995. [PMID: 38670255 DOI: 10.1016/j.drudis.2024.103995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Calcium ion dysregulation exerts profound effects on various physiological activities such as tumor proliferation, migration, and drug resistance. Calcium-related channels play a regulatory role in maintaining calcium ion homeostasis, with most channels being highly expressed in tumor cells. Additionally, these channels serve as potential drug targets for the development of antitumor medications. In this review, we first discuss the current research status of these pathways, examining how they modulate various tumor functions such as epithelial-mesenchymal transition (EMT), metabolism, and drug resistance. Simultaneously, we summarize the recent progress in the study of novel small-molecule drugs over the past 5 years and their current status.
Collapse
Affiliation(s)
- Linxi Zhang
- China Medical University-Queen's University of Belfast Joint College, China Medical University, Shenyang 110000, Liaoning, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu 611130, China
| | - Jiao Liu
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu 611130, China
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Chengyong Wu
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
5
|
Todesca LM, Gerke M, Bulk EE, Bachmann M, Rudersdorf A, Antonuzzo L, Pillozzi S, Düfer M, Szabo I, Schwab A. Targeting K Ca3.1 channels to overcome erlotinib resistance in non-small cell lung cancer cells. Cell Death Discov 2024; 10:2. [PMID: 38177097 PMCID: PMC10767088 DOI: 10.1038/s41420-023-01776-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Almost all non-small cell lung cancer (NSCLC) patients initially responding to EGFR tyrosine kinase inhibitors (TKIs) develop acquired resistance. Since KCa3.1 channels, expressed in mitochondria and plasma membrane, regulate similar behavioral traits of NSCLC cells as EGFR, we hypothesized that their blockade contributes to overcoming EGFR-TKI resistance. Meta-analysis of microarray data revealed that KCa3.1 channel expression in erlotinib-resistant NSCLC cells correlates with that of genes of integrin and apoptosis pathways. Using erlotinib-sensitive and -resistant NSCLC cells we monitored the role of mitochondrial KCa3.1 channels in integrin signaling by studying cell-matrix adhesion with single-cell force spectroscopy. Apoptosis was quantified with fluorescence-based assays. The function of mitochondrial KCa3.1 channels in these processes was assessed by measuring the mitochondrial membrane potential and by quantifying ROS production. Functional assays were supplemented by biochemical analyses. We show that KCa3.1 channel inhibition with senicapoc in erlotinib-resistant NSCLC cells increases cell adhesion by increasing β1-integrin expression, that in turn depends on mitochondrial ROS release. Increased adhesion impairs migration of NSCLC cells in a 3D matrix. At the same time, the senicapoc-dependent ROS production induces cytochrome C release and triggers apoptosis of erlotinib-resistant NSCLC cells. Thus, KCa3.1 channel blockade overcomes EGFR-TKI resistance by inhibiting NSCLC motility and inducing apoptosis.
Collapse
Affiliation(s)
| | - Matthias Gerke
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Emma Etmar Bulk
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | - Alisa Rudersdorf
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Martina Düfer
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
6
|
Cammann C, Kulla J, Wiebusch L, Walz C, Zhao F, Lowinus T, Topfstedt E, Mishra N, Henklein P, Bommhardt U, Bossaller L, Hagemeier C, Schadendorf D, Schmidt B, Paschen A, Seifert U. Proteasome inhibition potentiates Kv1.3 potassium channel expression as therapeutic target in drug-sensitive and -resistant human melanoma cells. Biomed Pharmacother 2023; 168:115635. [PMID: 37816303 DOI: 10.1016/j.biopha.2023.115635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/24/2023] [Accepted: 10/03/2023] [Indexed: 10/12/2023] Open
Abstract
Primary and acquired therapy resistance is a major problem in patients with BRAF-mutant melanomas being treated with BRAF and MEK inhibitors (BRAFI, MEKi). Therefore, development of alternative therapy regimes is still required. In this regard, new drug combinations targeting different pathways to induce apoptosis could offer promising alternative approaches. Here, we investigated the combination of proteasome and Kv1.3 potassium channel inhibition on chemo-resistant, BRAF inhibitor-resistant as well as sensitive human melanoma cells. Our experiments demonstrated that all analyzed melanoma cell lines were sensitive to proteasome inhibitor treatment at concentrations that are not toxic to primary human fibroblasts. To further reduce proteasome inhibitor-associated side effects, and to foster apoptosis, potassium channels, which are other targets to induce pro-apoptotic effects in cancer cells, were blocked. In support, combined exposure of melanoma cells to proteasome and Kv1.3 channel inhibitor resulted in synergistic effects and significantly reduced cell viability. On the molecular level, enhanced apoptosis correlated with an increase of intracellular Kv1.3 channels and pro-apoptotic proteins such as Noxa and Bak and a reduction of anti-apoptotic proteins. Thus, use of combined therapeutic strategies triggering different apoptotic pathways may efficiently prevent the outgrowth of drug-resistant and -sensitive BRAF-mutant melanoma cells. In addition, this could be the basis for an alternative approach to treat other tumors expressing mutated BRAF such as non-small-cell lung cancer.
Collapse
Affiliation(s)
- Clemens Cammann
- Friedrich Loeffler - Institute of Medical Microbiology - Virology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Jonas Kulla
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Lüder Wiebusch
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christian Walz
- Clemens Schöpf-Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Alarich Weiss-Straße 4-8, 64287 Darmstadt, Germany
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Eylin Topfstedt
- Friedrich Loeffler - Institute of Medical Microbiology - Virology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Neha Mishra
- Section of Rheumatology, Clinic and Policlinic of Internal Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Petra Henklein
- Institute of Molecular Biology and Biochemistry, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Lukas Bossaller
- Section of Rheumatology, Clinic and Policlinic of Internal Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Christian Hagemeier
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Boris Schmidt
- Clemens Schöpf-Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Alarich Weiss-Straße 4-8, 64287 Darmstadt, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Ulrike Seifert
- Friedrich Loeffler - Institute of Medical Microbiology - Virology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany.
| |
Collapse
|
7
|
Zuccolini P, Barbieri R, Sbrana F, Picco C, Gavazzo P, Pusch M. IK Channel-Independent Effects of Clotrimazole and Senicapoc on Cancer Cells Viability and Migration. Int J Mol Sci 2023; 24:16285. [PMID: 38003471 PMCID: PMC10671816 DOI: 10.3390/ijms242216285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Many studies highlighted the importance of the IK channel for the proliferation and the migration of different types of cancer cells, showing how IK blockers could slow down cancer growth. Based on these data, we wanted to characterize the effects of IK blockers on melanoma metastatic cells and to understand if such effects were exclusively IK-dependent. For this purpose, we employed two different blockers, namely clotrimazole and senicapoc, and two cell lines: metastatic melanoma WM266-4 and pancreatic cancer Panc-1, which is reported to have little or no IK expression. Clotrimazole and senicapoc induced a decrease in viability and the migration of both WM266-4 and Panc-1 cells irrespective of IK expression levels. Patch-clamp experiments on WM266-4 cells revealed Ca2+-dependent, IK-like, clotrimazole- and senicapoc-sensitive currents, which could not be detected in Panc-1 cells. Neither clotrimazole nor senicapoc altered the intracellular Ca2+ concentration. These results suggest that the effects of IK blockers on cancer cells are not strictly dependent on a robust presence of the channel in the plasma membrane, but they might be due to off-target effects on other cellular targets or to the blockade of IK channels localized in intracellular organelles.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Pusch
- Biophysics Institute, National Research Council, 16149 Genova, Italy; (P.Z.); (R.B.); (F.S.); (C.P.); (P.G.)
| |
Collapse
|
8
|
Capitani C, Chioccioli Altadonna G, Santillo M, Lastraioli E. Ion channels in lung cancer: biological and clinical relevance. Front Pharmacol 2023; 14:1283623. [PMID: 37942486 PMCID: PMC10627838 DOI: 10.3389/fphar.2023.1283623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023] Open
Abstract
Despite improvements in treatment, lung cancer is still a major health problem worldwide. Among lung cancer subtypes, the most frequent is represented by adenocarcinoma (belonging to the Non-Small Cell Lung Cancer class) although the most challenging and harder to treat is represented by Small Cell Lung Cancer, that occurs at lower frequency but has the worst prognosis. For these reasons, the standard of care for these patients is represented by a combination of surgery, radiation therapy and chemotherapy. In this view, searching for novel biomarkers that might help both in diagnosis and therapy is mandatory. In the last 30 years it was demonstrated that different families of ion channels are overexpressed in both lung cancer cell lines and primary tumours. The altered ion channel profile may be advantageous for diagnostic and therapeutic purposes since most of them are localised on the plasma membrane thus their detection is quite easy, as well as their block with specific drugs and antibodies. This review focuses on ion channels (Potassium, Sodium, Calcium, Chloride, Anion and Nicotinic Acetylcholine receptors) in lung cancer (both Non-Small Cell Lung Cancer and Small Cell Lung Cancer) and recapitulate the up-to-date knowledge about their role and clinical relevance for a potential use in the clinical setting, for lung cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Chiara Capitani
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ginevra Chioccioli Altadonna
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Michele Santillo
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Lastraioli
- General Pathology Laboratory, Department of Experimental and Clinical Medicine, Internal Medicine Section, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Chiliquinga AJ, Acosta B, Ogonaga-Borja I, Villarruel-Melquiades F, de la Garza J, Gariglio P, Ocádiz-Delgado R, Ramírez A, Sánchez-Pérez Y, García-Cuellar CM, Bañuelos C, Camacho J. Ion Channels as Potential Tools for the Diagnosis, Prognosis, and Treatment of HPV-Associated Cancers. Cells 2023; 12:1376. [PMID: 37408210 PMCID: PMC10217072 DOI: 10.3390/cells12101376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 07/07/2023] Open
Abstract
The human papilloma virus (HPV) group comprises approximately 200 genetic types that have a special affinity for epithelial tissues and can vary from producing benign symptoms to developing into complicated pathologies, such as cancer. The HPV replicative cycle affects various cellular and molecular processes, including DNA insertions and methylation and relevant pathways related to pRb and p53, as well as ion channel expression or function. Ion channels are responsible for the flow of ions across cell membranes and play very important roles in human physiology, including the regulation of ion homeostasis, electrical excitability, and cell signaling. However, when ion channel function or expression is altered, the channels can trigger a wide range of channelopathies, including cancer. In consequence, the up- or down-regulation of ion channels in cancer makes them attractive molecular markers for the diagnosis, prognosis, and treatment of the disease. Interestingly, the activity or expression of several ion channels is dysregulated in HPV-associated cancers. Here, we review the status of ion channels and their regulation in HPV-associated cancers and discuss the potential molecular mechanisms involved. Understanding the dynamics of ion channels in these cancers should help to improve early diagnosis, prognosis, and treatment in the benefit of HPV-associated cancer patients.
Collapse
Affiliation(s)
| | - Brenda Acosta
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Ingrid Ogonaga-Borja
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Fernanda Villarruel-Melquiades
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Jaime de la Garza
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Rodolfo Ocádiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Ana Ramírez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Calzada Universidad 14418, Tijuana 22390, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Claudia M. García-Cuellar
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| |
Collapse
|
10
|
Younes S, Mourad N, Salla M, Rahal M, Hammoudi Halat D. Potassium Ion Channels in Glioma: From Basic Knowledge into Therapeutic Applications. MEMBRANES 2023; 13:434. [PMID: 37103862 PMCID: PMC10144598 DOI: 10.3390/membranes13040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 06/19/2023]
Abstract
Ion channels, specifically those controlling the flux of potassium across cell membranes, have recently been shown to exhibit an important role in the pathophysiology of glioma, the most common primary central nervous system tumor with a poor prognosis. Potassium channels are grouped into four subfamilies differing by their domain structure, gating mechanisms, and functions. Pertinent literature indicates the vital functions of potassium channels in many aspects of glioma carcinogenesis, including proliferation, migration, and apoptosis. The dysfunction of potassium channels can result in pro-proliferative signals that are highly related to calcium signaling as well. Moreover, this dysfunction can feed into migration and metastasis, most likely by increasing the osmotic pressure of cells allowing the cells to initiate the "escape" and "invasion" of capillaries. Reducing the expression or channel blockage has shown efficacy in reducing the proliferation and infiltration of glioma cells as well as inducing apoptosis, priming several approaches to target potassium channels in gliomas pharmacologically. This review summarizes the current knowledge on potassium channels, their contribution to oncogenic transformations in glioma, and the existing perspectives on utilizing them as potential targets for therapy.
Collapse
Affiliation(s)
- Samar Younes
- Department of Biomedical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut 1103, Lebanon;
| | - Nisreen Mourad
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut 1103, Lebanon;
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
| | - Mohamed Salla
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Bekaa 146404, Lebanon;
| | - Mohamad Rahal
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
| | - Dalal Hammoudi Halat
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
- Academic Quality Department, QU Health, Qatar University, Doha 2713, Qatar;
| |
Collapse
|
11
|
Xia C, Liu C, Ren S, Cai Y, Zhang Q, Xia C. Potassium channels, tumorigenesis and targeted drugs. Biomed Pharmacother 2023; 162:114673. [PMID: 37031494 DOI: 10.1016/j.biopha.2023.114673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
Potassium channels play an important role in human physiological function. Recently, various molecular mechanisms have implicated abnormal functioning of potassium channels in the proliferation, migration, invasion, apoptosis, and cancer stem cell phenotype formation. Potassium channels also mediate the association of tumor cells with the tumor microenvironment. Meanwhile, potassium channels are important targets for cancer chemotherapy. A variety of drugs exert anti-cancer effects by modulating potassium channels in tumor cells. Therefore, there is a need to understand how potassium channels participate in tumor development and progression, which could reveal new, novel targets for cancer diagnosis and treatment. This review summarizes the roles of voltage-gated potassium channels, calcium-activated potassium channels, inwardly rectifying potassium channels, and two-pore domain potassium channels in tumorigenesis and the underlying mechanism of potassium channel-targeted drugs. Therefore, the study lays the foundation for rational and effective drug design and individualized clinical therapeutics.
Collapse
Affiliation(s)
- Cong Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China
| | - Can Liu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Shuangyi Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China
| | - Yantao Cai
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China
| | - Qianshi Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China.
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
12
|
Soret B, Hense J, Lüdtke S, Thale I, Schwab A, Düfer M. Pancreatic K Ca3.1 channels in health and disease. Biol Chem 2023; 404:339-353. [PMID: 36571487 DOI: 10.1515/hsz-2022-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/24/2022] [Indexed: 12/27/2022]
Abstract
Ion channels play an important role for regulation of the exocrine and the endocrine pancreas. This review focuses on the Ca2+-regulated K+ channel KCa3.1, encoded by the KCNN4 gene, which is present in both parts of the pancreas. In the islets of Langerhans, KCa3.1 channels are involved in the regulation of membrane potential oscillations characterizing nutrient-stimulated islet activity. Channel upregulation is induced by gluco- or lipotoxic conditions and might contribute to micro-inflammation and impaired insulin release in type 2 diabetes mellitus as well as to diabetes-associated renal and vascular complications. In the exocrine pancreas KCa3.1 channels are expressed in acinar and ductal cells. They are thought to play a role for anion secretion during digestion but their physiological role has not been fully elucidated yet. Pancreatic carcinoma, especially pancreatic ductal adenocarcinoma (PDAC), is associated with drastic overexpression of KCa3.1. For pharmacological targeting of KCa3.1 channels, we are discussing the possible benefits KCa3.1 channel inhibitors might provide in the context of diabetes mellitus and pancreatic cancer, respectively. We are also giving a perspective for the use of a fluorescently labeled derivative of the KCa3.1 blocker senicapoc as a tool to monitor channel distribution in pancreatic tissue. In summary, modulating KCa3.1 channel activity is a useful strategy for exo-and endocrine pancreatic disease but further studies are needed to evaluate its clinical suitability.
Collapse
Affiliation(s)
- Benjamin Soret
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Jurek Hense
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Simon Lüdtke
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Insa Thale
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Corrensstraße 48, D-48149 Münster, Germany
| | - Albrecht Schwab
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Martina Düfer
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
13
|
Chauhan G, Wang X, Yousry C, Gupta V. Scalable Production and In Vitro Efficacy of Inhaled Erlotinib Nanoemulsion for Enhanced Efficacy in Non-Small Cell Lung Cancer (NSCLC). Pharmaceutics 2023; 15:pharmaceutics15030996. [PMID: 36986858 PMCID: PMC10054254 DOI: 10.3390/pharmaceutics15030996] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a global concern as one of the leading causes of cancer deaths. The treatment options for NSCLC are limited to systemic chemotherapy, administered either orally or intravenously, with no local chemotherapies to target NSCLC. In this study, we have prepared nanoemulsions of tyrosine kinase inhibitor (TKI), erlotinib, using the single step, continuous manufacturing, and easily scalable hot melt extrusion (HME) technique without additional size reduction step. The formulated nanoemulsions were optimized and evaluated for their physiochemical properties, in vitro aerosol deposition behavior, and therapeutic activity against NSCLC cell lines both in vitro and ex vivo. The optimized nanoemulsion showed suitable aerosolization characteristics for deep lung deposition. The in vitro anti-cancer activity was tested against the NSCLC A549 cell line which exhibited 2.8-fold lower IC50 for erlotinib-loaded nanoemulsion, as compared to erlotinib-free solution. Furthermore, ex vivo studies using a 3D spheroid model also revealed higher efficacy of erlotinib-loaded nanoemulsion against NSCLC. Hence, inhalable nanoemulsion can be considered as a potential therapeutic approach for the local lung delivery of erlotinib to NSCLC.
Collapse
Affiliation(s)
- Gautam Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Xuechun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Carol Yousry
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo 11562, Egypt
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
14
|
Thale I, Maskri S, Grey L, Todesca LM, Budde T, Maisuls I, Strassert CA, Koch O, Schwab A, Wünsch B. Imaging of K Ca 3.1 Channels in Tumor Cells with PET and Small-Molecule Fluorescent Probes. ChemMedChem 2023; 18:e202200551. [PMID: 36315933 PMCID: PMC10098740 DOI: 10.1002/cmdc.202200551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/31/2022] [Indexed: 01/20/2023]
Abstract
The Ca2+ activated K+ channel KCa 3.1 is overexpressed in several human tumor cell lines, e. g. clear cell renal carcinoma, prostate cancer, non-small cell lung cancer. Highly aggressive cancer cells use this ion channel for key processes of the metastatic cascade such as migration, extravasation and invasion. Therefore, small molecules, which are able to image this KCa 3.1 channel in vitro and in vivo represent valuable diagnostic and prognostic tool compounds. The [18 F]fluoroethyltriazolyl substituted senicapoc was used as positron emission tomography (PET) tracer and showed promising properties for imaging of KCa 3.1 channels in lung adenocarcinoma cells in mice. The novel senicapoc BODIPY conjugates with two F-atoms (9 a) and with a F-atom and a methoxy moiety (9 b) at the B-atom led to the characteristic punctate staining pattern resulting from labeling of single KCa 3.1 channels in A549-3R cells. This punctate pattern was completely removed by preincubation with an excess of senicapoc confirming the high specificity of KCa 3.1 labeling. Due to the methoxy moiety at the B-atom and the additional oxyethylene unit in the spacer, 9 b exhibits higher polarity, which improves solubility and handling without reduction of fluorescence quantum yield. Docking studies using a cryo-electron microscopy (EM) structure of the KCa 3.1 channel confirmed the interaction of 9 a and 9 b with a binding pocket in the channel pore.
Collapse
Affiliation(s)
- Insa Thale
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Sarah Maskri
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Lucie Grey
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Luca Matteo Todesca
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology II, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Thomas Budde
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology I, Robert-Koch-Straße 27a, 48149, Münster, Germany
| | - Ivan Maisuls
- Westfälische Wilhelms-Universität Münster, Institut für Anorganische und Analytische Chemie CiMIC, SoN, Corrensstraße 28, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, CeNTech, Heisenbergstraße 11, 48149, Münster, Germany
| | - Cristian A Strassert
- Westfälische Wilhelms-Universität Münster, Institut für Anorganische und Analytische Chemie CiMIC, SoN, Corrensstraße 28, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, CeNTech, Heisenbergstraße 11, 48149, Münster, Germany
| | - Oliver Koch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Albrecht Schwab
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology II, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Bernhard Wünsch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| |
Collapse
|
15
|
Li M, Tian P, Zhao Q, Ma X, Zhang Y. Potassium channels: Novel targets for tumor diagnosis and chemoresistance. Front Oncol 2023; 12:1074469. [PMID: 36703789 PMCID: PMC9872028 DOI: 10.3389/fonc.2022.1074469] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
In recent years, the role of potassium channels in tumors has been intensively studied. Potassium channel proteins are widely involved in various physiological and pathological processes of cells. The expression and dysfunction of potassium channels are closely related to tumor progression. Potassium channel blockers or activators present antitumor effects by directly inhibiting tumor growth or enhancing the potency of classical antitumor agents in combination therapy. This article reviews the mechanisms by which potassium channels contribute to tumor development in various tumors in recent years, introduces the potential of potassium channels as diagnostic targets and therapeutic means for tumors, and provides further ideas for the proper individualized treatment of tumors.
Collapse
Affiliation(s)
- Meizeng Li
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Peijie Tian
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Qing Zhao
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Xialin Ma
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Yunxiang Zhang
- Department of Pathology, Weifang People’ s Hospital, Weifang, China,*Correspondence: Yunxiang Zhang,
| |
Collapse
|
16
|
IK Ca channels control breast cancer metabolism including AMPK-driven autophagy. Cell Death Dis 2022; 13:902. [PMID: 36302750 PMCID: PMC9613901 DOI: 10.1038/s41419-022-05329-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022]
Abstract
Ca2+-activated K+ channels of intermediate conductance (IK) are frequently overexpressed in breast cancer (BC) cells, while IK channel depletion reduces BC cell proliferation and tumorigenesis. This raises the question, of whether and mechanistically how IK activity interferes with the metabolic activity and energy consumption rates, which are fundamental for rapidly growing cells. Using BC cells obtained from MMTV-PyMT tumor-bearing mice, we show that both, glycolysis and mitochondrial ATP-production are reduced in cells derived from IK-deficient breast tumors. Loss of IK altered the sub-/cellular K+- and Ca2+- homeostasis and mitochondrial membrane potential, ultimately resulting in reduced ATP-production and metabolic activity. Consequently, we find that BC cells lacking IK upregulate AMP-activated protein kinase activity to induce autophagy compensating the glycolytic and mitochondrial energy shortage. Our results emphasize that IK by modulating cellular Ca2+- and K+-dynamics contributes to the remodeling of metabolic pathways in cancer. Thus, targeting IK channel might disturb the metabolic activity of BC cells and reduce malignancy.
Collapse
|
17
|
Fuest S, Post C, Balbach ST, Jabar S, Neumann I, Schimmelpfennig S, Sargin S, Nass E, Budde T, Kailayangiri S, Altvater B, Ranft A, Hartmann W, Dirksen U, Rössig C, Schwab A, Pethő Z. Relevance of Abnormal KCNN1 Expression and Osmotic Hypersensitivity in Ewing Sarcoma. Cancers (Basel) 2022; 14:4819. [PMID: 36230742 PMCID: PMC9564116 DOI: 10.3390/cancers14194819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 12/02/2022] Open
Abstract
Ewing sarcoma (EwS) is a rare and highly malignant bone tumor occurring mainly in childhood and adolescence. Physiologically, the bone is a central hub for Ca2+ homeostasis, which is severely disturbed by osteolytic processes in EwS. Therefore, we aimed to investigate how ion transport proteins involved in Ca2+ homeostasis affect EwS pathophysiology. We characterized the expression of 22 candidate genes of Ca2+-permeable or Ca2+-regulated ion channels in three EwS cell lines and found the Ca2+-activated K+ channel KCa2.1 (KCNN1) to be exceptionally highly expressed. We revealed that KCNN1 expression is directly regulated by the disease-driving oncoprotein EWSR1-FL1. Due to its consistent overexpression in EwS, KCNN1 mRNA could be a prognostic marker in EwS. In a large cohort of EwS patients, however, KCNN1 mRNA quantity does not correlate with clinical parameters. Several functional studies including patch clamp electrophysiology revealed no evidence for KCa2.1 function in EwS cells. Thus, elevated KCNN1 expression is not translated to KCa2.1 channel activity in EwS cells. However, we found that the low K+ conductance of EwS cells renders them susceptible to hypoosmotic solutions. The absence of a relevant K+ conductance in EwS thereby provides an opportunity for hypoosmotic therapy that can be exploited during tumor surgery.
Collapse
Affiliation(s)
- Sebastian Fuest
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Christoph Post
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Sebastian T Balbach
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Susanne Jabar
- Pediatrics III, University Hospital Essen, 45147 Essen, Germany
| | - Ilka Neumann
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | | | - Sarah Sargin
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Elke Nass
- Institute of Physiology I, University Münster, 48149 Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, University Münster, 48149 Münster, Germany
| | - Sareetha Kailayangiri
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Bianca Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Andreas Ranft
- Pediatrics III, University Hospital Essen, 45147 Essen, Germany
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, University Münster, 48149 Münster, Germany
| | - Uta Dirksen
- Pediatrics III, University Hospital Essen, 45147 Essen, Germany
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149 Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| | - Zoltán Pethő
- Institute of Physiology II, University Münster, 48149 Münster, Germany
| |
Collapse
|
18
|
Bulk E, Todesca LM, Bachmann M, Szabo I, Rieke M, Schwab A. Functional expression of mitochondrial K Ca3.1 channels in non-small cell lung cancer cells. Pflugers Arch 2022; 474:1147-1157. [PMID: 36152073 PMCID: PMC9560933 DOI: 10.1007/s00424-022-02748-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 09/05/2022] [Indexed: 12/01/2022]
Abstract
Lung cancer is one of the leading causes of cancer-related deaths worldwide. The Ca2+-activated K+ channel KCa3.1 contributes to the progression of non-small cell lung cancer (NSCLC). Recently, KCa3.1 channels were found in the inner membrane of mitochondria in different cancer cells. Mitochondria are the main sources for the generation of reactive oxygen species (ROS) that affect the progression of cancer cells. Here, we combined Western blotting, immunofluorescence, and fluorescent live-cell imaging to investigate the expression and function of KCa3.1 channels in the mitochondria of NSCLC cells. Western blotting revealed KCa3.1 expression in mitochondrial lysates from different NSCLC cells. Using immunofluorescence, we demonstrate a co-localization of KCa3.1 channels with mitochondria of NSCLC cells. Measurements of the mitochondrial membrane potential with TMRM reveal a hyperpolarization following the inhibition of KCa3.1 channels with the cell-permeable blocker senicapoc. This is not the case when cells are treated with the cell-impermeable peptidic toxin maurotoxin. The hyperpolarization of the mitochondrial membrane potential is accompanied by an increased generation of ROS in NSCLC cells. Collectively, our results provide firm evidence for the functional expression of KCa3.1 channels in the inner membrane of mitochondria of NSCLC cells.
Collapse
Affiliation(s)
- Etmar Bulk
- Institute of Physiology II, University of Münster, 48149, Münster, Germany.
| | | | | | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy
| | - Marius Rieke
- Institute of Physiology II, University of Münster, 48149, Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, 48149, Münster, Germany
| |
Collapse
|
19
|
Zúñiga L, Cayo A, González W, Vilos C, Zúñiga R. Potassium Channels as a Target for Cancer Therapy: Current Perspectives. Onco Targets Ther 2022; 15:783-797. [PMID: 35899081 PMCID: PMC9309325 DOI: 10.2147/ott.s326614] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/08/2022] [Indexed: 12/18/2022] Open
Abstract
Potassium (K+) channels are highly regulated membrane proteins that control the potassium ion flux and respond to different cellular stimuli. These ion channels are grouped into three major families, Kv (voltage-gated K+ channel), Kir (inwardly rectifying K+ channel) and K2P (two-pore K+ channels), according to the structure, to mediate the K+ currents. In cancer, alterations in K+ channel function can promote the acquisition of the so-called hallmarks of cancer – cell proliferation, resistance to apoptosis, metabolic changes, angiogenesis, and migratory capabilities – emerging as targets for the development of new therapeutic drugs. In this review, we focus our attention on the different K+ channels associated with the most relevant and prevalent cancer types. We summarize our knowledge about the potassium channels structure and function, their cancer dysregulated expression and discuss the K+ channels modulator and the strategies for designing new drugs.
Collapse
Affiliation(s)
- Leandro Zúñiga
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Angel Cayo
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca, Chile
| | - Cristian Vilos
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile.,Laboratory of Nanomedicine and Targeted Delivery, School of Medicine, Universidad de Talca, Talca, 3460000, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| | - Rafael Zúñiga
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| |
Collapse
|
20
|
Djamgoz MBA. Combinatorial Therapy of Cancer: Possible Advantages of Involving Modulators of Ionic Mechanisms. Cancers (Basel) 2022; 14:2703. [PMID: 35681682 PMCID: PMC9179511 DOI: 10.3390/cancers14112703] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer is a global health problem that 1 in 2-3 people can expect to experience during their lifetime. Several different modalities exist for cancer management, but all of these suffer from significant shortcomings in both diagnosis and therapy. Apart from developing completely new therapies, a viable way forward is to improve the efficacy of the existing modalities. One way is to combine these with each other or with other complementary approaches. An emerging latter approach is derived from ionic mechanisms, mainly ion channels and exchangers. We evaluate the evidence for this systematically for the main treatment methods: surgery, chemotherapy, radiotherapy and targeted therapies (including monoclonal antibodies, steroid hormones, tyrosine kinase inhibitors and immunotherapy). In surgery, the possible systemic use of local anesthetics to suppress subsequent relapse is still being discussed. For all the other methods, there is significant positive evidence for several cancers and a range of modulators of ionic mechanisms. This applies also to some of the undesirable side effects of the treatments. In chemotherapy, for example, there is evidence for co-treatment with modulators of the potassium channel (Kv11.1), pH regulation (sodium-hydrogen exchanger) and Na+-K+-ATPase (digoxin). Voltage-gated sodium channels, shown previously to promote metastasis, appear to be particularly useful for co-targeting with inhibitors of tyrosine kinases, especially epidermal growth factor. It is concluded that combining current orthodox treatment modalities with modulators of ionic mechanisms can produce beneficial effects including (i) making the treatment more effective, e.g., by lowering doses; (ii) avoiding the onset of resistance to therapy; (iii) reducing undesirable side effects. However, in many cases, prospective clinical trials are needed to put the findings firmly into clinical context.
Collapse
Affiliation(s)
- Mustafa B. A. Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; ; Tel.: +44-796-181-6959
- Biotechnology Research Centre, Cyprus International University, Haspolat, Mersin 10, Turkey
| |
Collapse
|