1
|
Rugi M, Hofschröer V, Pethő Z, Soret B, Loeck T, Schwab A. K 2P2.1 channels modulate the pH- and mechanosensitivity of pancreatic stellate cells. Pflugers Arch 2025; 477:147-157. [PMID: 39325089 PMCID: PMC11711774 DOI: 10.1007/s00424-024-03021-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
Pancreatic stellate cells (PSCs) are central in the development of acute pancreatitis and tumor fibrosis in pancreatic ductal adenocarcinoma (PDAC). Fibrosis and a unique pH landscape represent characteristic properties of the PDAC microenvironment. Mechanosensitive ion channels are involved in the activation of PSCs. Among these channels, K2P2.1 has not yet been studied in PSCs. K2P2.1 channels are pH- and mechanosensitive. We confirmed K2P2.1 expression in PSCs by RT-qPCR and immunofluorescence. PSCs from K2P2.1+/+ and K2P2.1-/- mice were studied under conditions mimicking properties of the PDAC microenvironment (acidic extracellular pH (pHe), ambient pressure elevated by + 100 mmHg). Migration and the cell area were taken as surrogates for PSC activation and evaluated with live cell imaging. pHe-dependent changes of the membrane potential of PSCs were investigated with DiBAC4(3), a voltage-sensitive fluorescent dye. We observed a correlation between morphological activation and progressive hyperpolarization of the cells in response to changes in pHe and pressure. The effect was in part dependent on the expression of K2P2.1 channels because the membrane potential of K2P2.1+/+ PSCs was always more hyperpolarized than that of K2P2.1-/- PSCs. Cell migration velocity of K2P2.1+/+ cells decreased upon pressure application when cells were kept in an acidic medium (pHe 6.6). This was not the case in K2P2.1-/- PSCs. Taken together, our study highlights the critical role of K2P2.1 channels in the combined sensing of environmental pressure and pHe by PSCs and in coordinating cellular morphology with membrane potential dynamics. Thus, K2P2.1 channels are important mechano-sensors in murine PSCs.
Collapse
Affiliation(s)
- Micol Rugi
- Institut Für Physiologie II, Robert-Koch-Str. 27B, 48149, Münster, Germany
| | - Verena Hofschröer
- Institut Für Physiologie II, Robert-Koch-Str. 27B, 48149, Münster, Germany
| | - Zoltán Pethő
- Institut Für Physiologie II, Robert-Koch-Str. 27B, 48149, Münster, Germany
| | - Benjamin Soret
- Institut Für Physiologie II, Robert-Koch-Str. 27B, 48149, Münster, Germany
- Laboratory of Cell Physiology, INSERM U 1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - Thorsten Loeck
- Institut Für Physiologie II, Robert-Koch-Str. 27B, 48149, Münster, Germany
| | - Albrecht Schwab
- Institut Für Physiologie II, Robert-Koch-Str. 27B, 48149, Münster, Germany.
| |
Collapse
|
2
|
Deshar G, Christensen NM, Novak I. Pantoprazole and riluzole target H +/K +-ATPases and pH-sensitive K + channels in pancreatic cancer cells. Int J Cancer 2024; 155:1641-1654. [PMID: 38975879 DOI: 10.1002/ijc.35076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/28/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains the most lethal cancer type. PDAC is characterized by fibrotic, hypoxic, and presumably acidic tumor microenvironment (TME). Acidic TME is an important player in tumor development, progression, aggressiveness, and chemoresistance. The dysregulation of ductal ion transporters/channels might contribute to extracellular pH (pHe) acidification and PDAC progression. Our aim was to test whether H+/K+-ATPases and pH-sensitive K+ channels contribute to these processes and could be targeted by clinically approved drugs. We used human pancreatic cancer cells adapted to various pHe conditions and grown in monolayers and spheroids. First, we created cells expressing pHoran4 at the outer plasma membrane and showed that pantoprazole, the H+/K+-ATPase inhibitor, alkalinized pHe. Second, we used FluoVolt to monitor the membrane voltage (Vm) and showed that riluzole hyperpolarized Vm, most likely by opening of pH-sensitive K+ channels such as TREK-1. Third, we show that pantoprazole and riluzole inhibited cell proliferation and viability of monolayers and spheroids of cancer cells adapted to various pHe conditions. Most importantly, combination of the two drugs had significantly larger inhibitory effects on PDAC cell survival. We propose that co-targeting H+/K+-ATPases and pH-sensitive K+ channels by re-purposing of pantoprazole and riluzole could provide novel acidosis-targeted therapies of PDAC.
Collapse
Affiliation(s)
- Ganga Deshar
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Ivana Novak
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Piga M, Varga Z, Feher A, Papp F, Korpos E, Bangera KC, Frlan R, Ilaš J, Dernovšek J, Tomašič T, Zidar N. Identification of a Novel Structural Class of H V1 Inhibitors by Structure-Based Virtual Screening. J Chem Inf Model 2024; 64:4850-4862. [PMID: 38850237 PMCID: PMC11200261 DOI: 10.1021/acs.jcim.4c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
The human voltage-gated proton channel, hHV1, is highly expressed in various cell types including macrophages, B lymphocytes, microglia, sperm cells and also in various cancer cells. Overexpression of HV1 has been shown to promote tumor formation by highly metastatic cancer cells, and has been associated with neuroinflammatory diseases, immune response disorders and infertility, suggesting a potential use of hHV1 inhibitors in numerous therapeutic areas. To identify compounds targeting this channel, we performed a structure-based virtual screening on an open structure of the human HV1 channel. Twenty selected virtual screening hits were tested on Chinese hamster ovary (CHO) cells transiently expressing hHV1, with compound 13 showing strong block of the proton current with an IC50 value of 8.5 μM. Biological evaluation of twenty-three additional analogs of 13 led to the discovery of six other compounds that blocked the proton current by more than 50% at 50 μM concentration. This allowed for an investigation of structure-activity relationships. The antiproliferative activity of the selected promising hHV1 inhibitors was investigated in the cell lines MDA-MB-231 and THP-1, where compound 13 inhibited growth with an IC50 value of 9.0 and 8.1 μM, respectively. The identification of a new structural class of HV1 inhibitors contributes to our understanding of the structural requirements for inhibition of this ion channel and opens up the possibility of investigating the role of HV1 inhibitors in various pathological conditions and in cancer therapy.
Collapse
Affiliation(s)
- Martina Piga
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Zoltan Varga
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Adam Feher
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Ferenc Papp
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Eva Korpos
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
- HUN-REN−UD
Cell Biology and Signaling Research Group, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Kavya C. Bangera
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Rok Frlan
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Janez Ilaš
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Jaka Dernovšek
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Tihomir Tomašič
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Nace Zidar
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| |
Collapse
|
4
|
Cao Y, Wong RCH, Xue EY, Zhang H, Wang J, Ding Y, Zhang L, Chen F, Wang J, Ng DKP. Immobilising an acid-cleavable dimeric phthalocyanine on gold nanobipyramids for intracellular pH detection and photodynamic elimination of cancer cells. Analyst 2024; 149:3288-3292. [PMID: 38808583 DOI: 10.1039/d4an00420e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
An acetal-linked dimeric phthalocyanine has been synthesised and immobilised on the surface of gold nanobipyramids. The resulting nanocomposite serves as a highly sensitive probe for intracellular pH through its acid-responsive fluorescence and surface-enhanced Raman scattering signals. The phthalocyanine units released in the acidic intracellular environment can also effectively eliminate the cancer cells upon light irradiation, rendering this simple fabricated nanosystem a bimodal and bifunctional theranostic agent.
Collapse
Affiliation(s)
- Yue Cao
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Roy C H Wong
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Evelyn Y Xue
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Han Zhang
- Department of Physics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Jie Wang
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yan Ding
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Lei Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials, Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| |
Collapse
|
5
|
Wang Q, Li H, Wu T, Yu B, Cong H, Shen Y. Nanodrugs based on co-delivery strategies to combat cisplatin resistance. J Control Release 2024; 370:14-42. [PMID: 38615892 DOI: 10.1016/j.jconrel.2024.04.020] [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: 12/30/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Qiubo Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Taixia Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bio-nanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
6
|
Stock C. pH-regulated single cell migration. Pflugers Arch 2024; 476:639-658. [PMID: 38214759 PMCID: PMC11006768 DOI: 10.1007/s00424-024-02907-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Over the last two decades, extra- and intracellular pH have emerged as fundamental regulators of cell motility. Fundamental physiological and pathological processes relying on appropriate cell migration, such as embryonic development, wound healing, and a proper immune defense on the one hand, and autoimmune diseases, metastatic cancer, and the progression of certain parasitic diseases on the other, depend on surrounding pH. In addition, migrating single cells create their own localized pH nanodomains at their surface and in the cytosol. By this means, the migrating cells locally modulate their adhesion to, and the re-arrangement and digestion of, the extracellular matrix. At the same time, the cytosolic nanodomains tune cytoskeletal dynamics along the direction of movement resulting in concerted lamellipodia protrusion and rear end retraction. Extracellular pH gradients as found in wounds, inflamed tissues, or the periphery of tumors stimulate directed cell migration, and long-term exposure to acidic conditions can engender a more migratory and invasive phenotype persisting for hours up to several generations of cells after they have left the acidic milieu. In the present review, the different variants of pH-dependent single cell migration are described. The underlying pH-dependent molecular mechanisms such as conformational changes of adhesion molecules, matrix protease activity, actin (de-)polymerization, and signaling events are explained, and molecular pH sensors stimulated by H+ signaling are presented.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hepatology, Infectiology & Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
7
|
DeCoursey TE. Transcendent Aspects of Proton Channels. Annu Rev Physiol 2024; 86:357-377. [PMID: 37931166 PMCID: PMC10938948 DOI: 10.1146/annurev-physiol-042222-023242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
A handful of biological proton-selective ion channels exist. Some open at positive or negative membrane potentials, others open at low or high pH, and some are light activated. This review focuses on common features that result from the unique properties of protons. Proton conduction through water or proteins differs qualitatively from that of all other ions. Extraordinary proton selectivity is needed to ensure that protons permeate and other ions do not. Proton selectivity arises from a proton pathway comprising a hydrogen-bonded chain that typically includes at least one titratable amino acid side chain. The enormously diverse functions of proton channels in disparate regions of the phylogenetic tree can be summarized by considering the chemical and electrical consequences of proton flux across membranes. This review discusses examples of cells in which proton efflux serves to increase pHi, decrease pHo, control the membrane potential, generate action potentials, or compensate transmembrane movement of electrical charge.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois, USA;
| |
Collapse
|
8
|
Pethő Z, Najder K, Beel S, Fels B, Neumann I, Schimmelpfennig S, Sargin S, Wolters M, Grantins K, Wardelmann E, Mitkovski M, Oeckinghaus A, Schwab A. Acid-base homeostasis orchestrated by NHE1 defines the pancreatic stellate cell phenotype in pancreatic cancer. JCI Insight 2023; 8:e170928. [PMID: 37643024 PMCID: PMC10619433 DOI: 10.1172/jci.insight.170928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) progresses in an organ with a unique pH landscape, where the stroma acidifies after each meal. We hypothesized that disrupting this pH landscape during PDAC progression triggers pancreatic stellate cells (PSCs) and cancer-associated fibroblasts (CAFs) to induce PDAC fibrosis. We revealed that alkaline environmental pH was sufficient to induce PSC differentiation to a myofibroblastic phenotype. We then mechanistically dissected this finding, focusing on the involvement of the Na+/H+ exchanger NHE1. Perturbing cellular pH homeostasis by inhibiting NHE1 with cariporide partially altered the myofibroblastic PSC phenotype. To show the relevance of this finding in vivo, we targeted NHE1 in murine PDAC (KPfC). Indeed, tumor fibrosis decreased when mice received the NHE1-inhibitor cariporide in addition to gemcitabine treatment. Moreover, the tumor immune infiltrate shifted from granulocyte rich to more lymphocytic. Taken together, our study provides mechanistic evidence on how the pancreatic pH landscape shapes pancreatic cancer through tuning PSC differentiation.
Collapse
Affiliation(s)
| | | | - Stephanie Beel
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Benedikt Fels
- Institute of Physiology II and
- Institute of Physiology, University of Lübeck, Lübeck, Germany
| | | | | | | | - Maria Wolters
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | - Klavs Grantins
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Andrea Oeckinghaus
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | | |
Collapse
|
9
|
Malcolm JR, Sajjaboontawee N, Yerlikaya S, Plunkett-Jones C, Boxall PJ, Brackenbury WJ. Voltage-gated sodium channels, sodium transport and progression of solid tumours. CURRENT TOPICS IN MEMBRANES 2023; 92:71-98. [PMID: 38007270 DOI: 10.1016/bs.ctm.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Sodium (Na+) concentration in solid tumours of different origin is highly dysregulated, and this corresponds to the aberrant expression of Na+ transporters. In particular, the α subunits of voltage gated Na+ channels (VGSCs) raise intracellular Na+ concentration ([Na+]i) in malignant cells, which influences the progression of solid tumours, predominantly driving cancer cells towards a more aggressive and metastatic phenotype. Conversely, re-expression of VGSC β subunits in cancer cells can either enhance tumour progression or promote anti-tumourigenic properties. Metastasis is the leading cause of cancer-related mortality, highlighting an important area of research which urgently requires improved therapeutic interventions. Here, we review the extent to which VGSC subunits are dysregulated in solid tumours, and consider the implications of such dysregulation on solid tumour progression. We discuss current understanding of VGSC-dependent mechanisms underlying increased invasive and metastatic potential of solid tumours, and how the complex relationship between the tumour microenvironment (TME) and VGSC expression may further drive tumour progression, in part due to the interplay of infiltrating immune cells, cancer-associated fibroblasts (CAFs) and insufficient supply of oxygen (hypoxia). Finally, we explore past and present clinical trials that investigate utilising existing VGSC modulators as potential pharmacological options to support adjuvant chemotherapies to prevent cancer recurrence. Such research demonstrates an exciting opportunity to repurpose therapeutics in order to improve the disease-free survival of patients with aggressive solid tumours.
Collapse
Affiliation(s)
- Jodie R Malcolm
- Department of Biology, University of York, Heslington, York, United Kingdom
| | - Nattanan Sajjaboontawee
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom
| | - Serife Yerlikaya
- Department of Biology, University of York, Heslington, York, United Kingdom; Istanbul Medipol University, Research Institute for Health Sciences and Technologies, Istanbul, Turkey
| | | | - Peter J Boxall
- Department of Biology, University of York, Heslington, York, United Kingdom; York and Scarborough Teaching Hospitals NHS Foundation Trust, York, United Kingdom
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom.
| |
Collapse
|
10
|
Loeck T, Rugi M, Todesca LM, Kalinowska P, Soret B, Neumann I, Schimmelpfennig S, Najder K, Pethő Z, Farfariello V, Prevarskaya N, Schwab A. The context-dependent role of the Na +/Ca 2+-exchanger (NCX) in pancreatic stellate cell migration. Pflugers Arch 2023; 475:1225-1240. [PMID: 37566113 PMCID: PMC10499968 DOI: 10.1007/s00424-023-02847-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 06/16/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023]
Abstract
Pancreatic stellate cells (PSCs) that can co-metastasize with cancer cells shape the tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) by producing an excessive amount of extracellular matrix. This leads to a TME characterized by increased tissue pressure, hypoxia, and acidity. Moreover, cells within the tumor secrete growth factors. The stimuli of the TME trigger Ca2+ signaling and cellular Na+ loading. The Na+/Ca2+ exchanger (NCX) connects the cellular Ca2+ and Na+ homeostasis. The NCX is an electrogenic transporter, which shuffles 1 Ca2+ against 3 Na+ ions over the plasma membrane in a forward or reverse mode. Here, we studied how the impact of NCX activity on PSC migration is modulated by cues from the TME. NCX expression was revealed with qPCR and Western blot. [Ca2+]i, [Na+]i, and the cell membrane potential were determined with the fluorescent indicators Fura-2, Asante NaTRIUM Green-2, and DiBAC4(3), respectively. PSC migration was quantified with live-cell imaging. To mimic the TME, PSCs were exposed to hypoxia, pressure, acidic pH (pH 6.6), and PDGF. NCX-dependent signaling was determined with Western blot analyses. PSCs express NCX1.3 and NCX1.9. [Ca2+]i, [Na+]i, and the cell membrane potential are 94.4 nmol/l, 7.4 mmol/l, and - 39.8 mV, respectively. Thus, NCX1 usually operates in the forward (Ca2+ export) mode. NCX1 plays a differential role in translating cues from the TME into an altered migratory behavior. When NCX1 is operating in the forward mode, its inhibition accelerates PSC migration. Thus, NCX1-mediated extrusion of Ca2+ contributes to a slow mode of migration of PSCs.
Collapse
Affiliation(s)
- Thorsten Loeck
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Micol Rugi
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Luca Matteo Todesca
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Paulina Kalinowska
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Benjamin Soret
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
- Université de Lille, Inserm, U1003 - PhyCell - Physiologie Cellulaire, F-59000, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Ilka Neumann
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Sandra Schimmelpfennig
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Karolina Najder
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Zoltán Pethő
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Valerio Farfariello
- Université de Lille, Inserm, U1003 - PhyCell - Physiologie Cellulaire, F-59000, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Université de Lille, Inserm, U1003 - PhyCell - Physiologie Cellulaire, F-59000, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Robert-Koch-Straße 27b, 48149, Münster, Germany.
| |
Collapse
|
11
|
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
|
12
|
Loeck T, Schwab A. The role of the Na +/Ca 2+-exchanger (NCX) in cancer-associated fibroblasts. Biol Chem 2023; 404:325-337. [PMID: 36594183 DOI: 10.1515/hsz-2022-0253] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/09/2022] [Indexed: 01/04/2023]
Abstract
Cancer is characterized by uncontrolled growth, invasion, and metastasis. In addition to solid cancer cells, cancer-associated fibroblasts (CAFs) play important roles in cancer pathophysiology. They arise from "healthy" cells but get manipulated by solid cancer cells to supply them and develop a tumor microenvironment (TME) that protects the cancer cells from the immune defense. A wide variety of cell types can differentiate into CAFs, including fibroblasts, endothelial cells, and epithelial cells. Precise Ca2+ regulation is essential for each cell including CAFs. The electrogenic Na+/Ca2+ exchanger (NCX) is one of the ubiquitously expressed regulatory Ca2+ transport proteins that rapidly responds to changes of the intracellular ion concentrations. Its transport function is also influenced by the membrane potential and thereby indirectly by the activity of ion channels. NCX transports Ca2+ out of the cell (forward mode) or allows its influx (reverse mode), always in exchange for 3 Na+ ions that are moved into the opposite direction. In this review, we discuss the functional roles NCX has in CAFs and how these depend on the properties of the TME. NCX activity modifies migration and leads to a reduced proliferation and apoptosis. The effect of the NCX in fibrosis is still largely unknown.
Collapse
Affiliation(s)
- Thorsten Loeck
- Institut für Physiologie II, Westfälische Wilhelms-Universität Münster, Robert-Koch-Str. 27b, D-48149 Münster, Germany
| | - Albrecht Schwab
- Institut für Physiologie II, Westfälische Wilhelms-Universität Münster, Robert-Koch-Str. 27b, D-48149 Münster, Germany
| |
Collapse
|
13
|
Cozzolino M, Gyöngyösi A, Korpos E, Gogolak P, Naseem MU, Kállai J, Lanyi A, Panyi G. The Voltage-Gated Hv1 H+ Channel Is Expressed in Tumor-Infiltrating Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:ijms24076216. [PMID: 37047188 PMCID: PMC10094655 DOI: 10.3390/ijms24076216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key determinants of the immunosuppressive microenvironment in tumors. As ion channels play key roles in the physiology/pathophysiology of immune cells, we aimed at studying the ion channel repertoire in tumor-derived polymorphonuclear (PMN-MDSC) and monocytic (Mo-MDSC) MDSCs. Subcutaneous tumors in mice were induced by the Lewis lung carcinoma cell line (LLC). The presence of PMN-MDSC (CD11b+/Ly6G+) and Mo-MDSCs (CD11b+/Ly6C+) in the tumor tissue was confirmed using immunofluorescence microscopy and cells were identified as CD11b+/Ly6G+ PMN-MDSCs and CD11b+/Ly6C+/F4/80−/MHCII− Mo-MDSCs using flow cytometry and sorting. The majority of the myeloid cells infiltrating the LLC tumors were PMN-MDSC (~60%) as compared to ~10% being Mo-MDSCs. We showed that PMN- and Mo-MDSCs express the Hv1 H+ channel both at the mRNA and at the protein level and that the biophysical and pharmacological properties of the whole-cell currents recapitulate the hallmarks of Hv1 currents: ~40 mV shift in the activation threshold of the current per unit change in the extracellular pH, high H+ selectivity, and sensitivity to the Hv1 inhibitor ClGBI. As MDSCs exert immunosuppression mainly by producing reactive oxygen species which is coupled to Hv1-mediated H+ currents, Hv1 might be an attractive target for inhibition of MDSCs in tumors.
Collapse
Affiliation(s)
- Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
| | - Adrienn Gyöngyösi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Eva Korpos
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Peter Gogolak
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Muhammad Umair Naseem
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
| | - Judit Kállai
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Arpad Lanyi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
- Correspondence: ; Tel.: +36-52-352201
| |
Collapse
|
14
|
Hu S, Cheng Q, Shang Y, Wang Z, Zhu R, Zhang L, Wu W, Zhang S, Li J. Synthesis of pH-responsive polyzwitterions for activated cellular uptake and tumor accumulation of gold nanoparticles at tumorous acidity. Biomed Mater 2023; 18. [PMID: 36645918 DOI: 10.1088/1748-605x/acb394] [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: 09/14/2022] [Accepted: 01/16/2023] [Indexed: 01/18/2023]
Abstract
The response sensitivity of surface material plays an important role in adjustable nano-bio interactionin vivo. In this present, a zwitterionic polymer (polyzwitterion) containing quaternary ammonium cation and sulfonamide anion poly(4-((4-(3-(methacryloyloxy)propoxy)phenyl) sulfonamido)-N, N, N-trimethyl-4-oxobutan-1-aminium chloride) (PMPTSA) was synthesized by Reversible Addition-Fragmentation Chain Transfer Polymerization (RAFT) polymerization to explore the pH responsive behavior in tumors. The PMPTSA-coated gold nanoparticles (PMPTSA-@-Au NPs) showed zwitterionic nature such as antifouling ability, low cellular uptake and prolonged circulation time similar with common hydrophilic polymers, including polyethylene glycol (PEG), poly(carboxybetaine methacrylate) and poly(sulfobetaine methacrylate) functional gold nanoparticles in physiological environment (pH 7.4). A high sensitivity and reversible positive charge conversion of P(MPTSA)-@-Au NPs at tumor slight acidic microenvironment (∼pH 6.8) leaded to an enhanced cellular internalization than that at pH 7.4 and increased tumor accumulation compared with PEG, polycarboxybetaines and polymer sulphobetaine (PSB) functional gold nanoparticles. The highly pH responsive PMPTSA will provide the promising application in cancer nanomedicine.
Collapse
Affiliation(s)
- Shumin Hu
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Qiuli Cheng
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Yulu Shang
- 989 Hospital of Joint Service Support Force of Chinese Pla, Luoyang 471023, People's Republic of China
| | - Zhihao Wang
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Rui Zhu
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Leitao Zhang
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Wenlan Wu
- School of Medicine, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| | - Shouren Zhang
- Henan Provincial Key Laboratory of Nanocomposite and Applications Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, Henan 450006, People's Republic of China
| | - Junbo Li
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, People's Republic of China
| |
Collapse
|
15
|
Feher A, Pethő Z, Szanto TG, Klekner Á, Tajti G, Batta G, Hortobágyi T, Varga Z, Schwab A, Panyi G. Mapping the functional expression of auxiliary subunits of K Ca1.1 in glioblastoma. Sci Rep 2022; 12:22023. [PMID: 36539587 PMCID: PMC9768140 DOI: 10.1038/s41598-022-26196-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive glial tumor, where ion channels, including KCa1.1, are candidates for new therapeutic options. Since the auxiliary subunits linked to KCa1.1 in GBM are largely unknown we used electrophysiology combined with pharmacology and gene silencing to address the functional expression of KCa1.1/β subunits complexes in both primary tumor cells and in the glioblastoma cell line U-87 MG. The pattern of the sensitivity (activation/inhibition) of the whole-cell currents to paxilline, lithocholic acid, arachidonic acid, and iberiotoxin; the presence of inactivation of the whole-cell current along with the loss of the outward rectification upon exposure to the reducing agent DTT collectively argue that KCa1.1/β3 complex is expressed in U-87 MG. Similar results were found using human primary glioblastoma cells isolated from patient samples. Silencing the β3 subunit expression inhibited carbachol-induced Ca2+ transients in U-87 MG thereby indicating the role of the KCa1.1/β3 in the Ca2+ signaling of glioblastoma cells. Functional expression of the KCa1.1/β3 complex, on the other hand, lacks cell cycle dependence. We suggest that the KCa1.1/β3 complex may have diagnostic and therapeutic potential in glioblastoma in the future.
Collapse
Affiliation(s)
- Adam Feher
- Department of Biophysics and Cell Biology, Faculty of Medicine, University Debrecen, Debrecen, Hungary
| | - Zoltán Pethő
- Department of Biophysics and Cell Biology, Faculty of Medicine, University Debrecen, Debrecen, Hungary
- Institute of Physiology II, University Münster, Münster, Germany
| | - Tibor G Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, University Debrecen, Debrecen, Hungary
| | - Álmos Klekner
- Department of Neurosurgery, Faculty of Medicine, University Debrecen, Debrecen, Hungary
| | - Gabor Tajti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University Debrecen, Debrecen, Hungary
| | - Gyula Batta
- Department of Genetics and Applied Microbiology, University Debrecen, Debrecen, Hungary
| | - Tibor Hortobágyi
- Faculty of Medicine, Institute of Pathology, University of Szeged, Szeged, Hungary
- ELKH-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, Faculty of Medicine, University Debrecen, Debrecen, Hungary
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University Debrecen, Debrecen, Hungary
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, Münster, Germany
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University Debrecen, Debrecen, Hungary.
| |
Collapse
|
16
|
Di Gregorio E, Israel S, Staelens M, Tankel G, Shankar K, Tuszyński JA. The distinguishing electrical properties of cancer cells. Phys Life Rev 2022; 43:139-188. [PMID: 36265200 DOI: 10.1016/j.plrev.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
In recent decades, medical research has been primarily focused on the inherited aspect of cancers, despite the reality that only 5-10% of tumours discovered are derived from genetic causes. Cancer is a broad term, and therefore it is inaccurate to address it as a purely genetic disease. Understanding cancer cells' behaviour is the first step in countering them. Behind the scenes, there is a complicated network of environmental factors, DNA errors, metabolic shifts, and electrostatic alterations that build over time and lead to the illness's development. This latter aspect has been analyzed in previous studies, but how the different electrical changes integrate and affect each other is rarely examined. Every cell in the human body possesses electrical properties that are essential for proper behaviour both within and outside of the cell itself. It is not yet clear whether these changes correlate with cell mutation in cancer cells, or only with their subsequent development. Either way, these aspects merit further investigation, especially with regards to their causes and consequences. Trying to block changes at various levels of occurrence or assisting in their prevention could be the key to stopping cells from becoming cancerous. Therefore, a comprehensive understanding of the current knowledge regarding the electrical landscape of cells is much needed. We review four essential electrical characteristics of cells, providing a deep understanding of the electrostatic changes in cancer cells compared to their normal counterparts. In particular, we provide an overview of intracellular and extracellular pH modifications, differences in ionic concentrations in the cytoplasm, transmembrane potential variations, and changes within mitochondria. New therapies targeting or exploiting the electrical properties of cells are developed and tested every year, such as pH-dependent carriers and tumour-treating fields. A brief section regarding the state-of-the-art of these therapies can be found at the end of this review. Finally, we highlight how these alterations integrate and potentially yield indications of cells' malignancy or metastatic index.
Collapse
Affiliation(s)
- Elisabetta Di Gregorio
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Simone Israel
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Michael Staelens
- Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada
| | - Gabriella Tankel
- Department of Mathematics & Statistics, McMaster University, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | - Karthik Shankar
- Department of Electrical & Computer Engineering, University of Alberta, 9211 116 Street NW, Edmonton, T6G 1H9, AB, Canada
| | - Jack A Tuszyński
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada; Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, T6G 1Z2, AB, Canada.
| |
Collapse
|
17
|
Papp F, Toombes GES, Pethő Z, Bagosi A, Feher A, Almássy J, Borrego J, Kuki Á, Kéki S, Panyi G, Varga Z. Multiple mechanisms contribute to fluorometry signals from the voltage-gated proton channel. Commun Biol 2022; 5:1131. [PMID: 36289443 PMCID: PMC9606259 DOI: 10.1038/s42003-022-04065-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Voltage-clamp fluorometry (VCF) supplies information about the conformational changes of voltage-gated proteins. Changes in the fluorescence intensity of the dye attached to a part of the protein that undergoes a conformational rearrangement upon the alteration of the membrane potential by electrodes constitute the signal. The VCF signal is generated by quenching and dequenching of the fluorescence as the dye traverses various local environments. Here we studied the VCF signal generation, using the Hv1 voltage-gated proton channel as a tool, which shares a similar voltage-sensor structure with voltage-gated ion channels but lacks an ion-conducting pore. Using mutagenesis and lipids added to the extracellular solution we found that the signal is generated by the combined effects of lipids during movement of the dye relative to the plane of the membrane and by quenching amino acids. Our 3-state model recapitulates the VCF signals of the various mutants and is compatible with the accepted model of two major voltage-sensor movements. Fluorometry signals indicating conformational change in an ion channel are generated by quenching amino acids and lipid effects during movement of the dye relative to the plane of the membrane.
Collapse
Affiliation(s)
- Ferenc Papp
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Gilman E. S. Toombes
- grid.94365.3d0000 0001 2297 5165Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Dr., MSC 3701, Bethesda, MD 20892-3701 USA
| | - Zoltán Pethő
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary ,grid.5949.10000 0001 2172 9288Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Adrienn Bagosi
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Adam Feher
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - János Almássy
- grid.7122.60000 0001 1088 8582Department of Physiology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Jesús Borrego
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Ákos Kuki
- grid.7122.60000 0001 1088 8582Department of Applied Chemistry, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Sándor Kéki
- grid.7122.60000 0001 1088 8582Department of Applied Chemistry, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Gyorgy Panyi
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| | - Zoltan Varga
- grid.7122.60000 0001 1088 8582Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen, H-4032 Hungary
| |
Collapse
|
18
|
Acid Adaptation Promotes TRPC1 Plasma Membrane Localization Leading to Pancreatic Ductal Adenocarcinoma Cell Proliferation and Migration through Ca 2+ Entry and Interaction with PI3K/CaM. Cancers (Basel) 2022; 14:cancers14194946. [PMID: 36230869 PMCID: PMC9563726 DOI: 10.3390/cancers14194946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers globally, with a 5-year overall survival of less than 10%. The development and progression of PDAC are linked to its fluctuating acidic tumor microenvironment. Ion channels act as important sensors of this acidic tumor microenvironment. They transduce extracellular signals and regulate signaling pathways involved in all hallmarks of cancer. In this study, we evaluated the interplay between a pH-sensitive ion channel, the calcium (Ca2+) channel transient receptor potential C1 (TRPC1), and three different stages of the tumor microenvironment, normal pH, acid adaptation, and acid recovery, and its impact on PDAC cell migration, proliferation, and cell cycle progression. In acid adaptation and recovery conditions, TRPC1 localizes to the plasma membrane, where it interacts with PI3K and calmodulin, and permits Ca2+ entry, which results in downstream signaling, leading to proliferation and migration. Thus, TRPC1 exerts a more aggressive role after adaptation to the acidic tumor microenvironment. Abstract Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a low overall survival rate of less than 10% and limited therapeutic options. Fluctuations in tumor microenvironment pH are a hallmark of PDAC development and progression. Many ion channels are bona fide cellular sensors of changes in pH. Yet, the interplay between the acidic tumor microenvironment and ion channel regulation in PDAC is poorly understood. In this study, we show that acid adaption increases PANC-1 cell migration but attenuates proliferation and spheroid growth, which are restored upon recovery. Moreover, acid adaptation and recovery conditions favor the plasma membrane localization of the pH-sensitive calcium (Ca2+) channel transient receptor potential C1 (TRPC1), TRPC1-mediated Ca2+ influx, channel interaction with the PI3K p85α subunit and calmodulin (CaM), and AKT and ERK1/2 activation. Knockdown (KD) of TRPC1 suppresses cell migration, proliferation, and spheroid growth, notably in acid-recovered cells. KD of TRPC1 causes the accumulation of cells in G0/G1 and G2/M phases, along with reduced expression of CDK6, −2, and −1, and cyclin A, and increased expression of p21CIP1. TRPC1 silencing decreases the basal Ca2+ influx in acid-adapted and -recovered cells, but not in normal pH conditions, and Ca2+ chelation reduces cell migration and proliferation solely in acid adaptation and recovery conditions. In conclusion, acid adaptation and recovery reinforce the involvement of TRPC1 in migration, proliferation, and cell cycle progression by permitting Ca2+ entry and forming a complex with the PI3K p85α subunit and CaM.
Collapse
|
19
|
Luis E, Anaya-Hernández A, León-Sánchez P, Durán-Pastén ML. The Kv10.1 Channel: A Promising Target in Cancer. Int J Mol Sci 2022; 23:ijms23158458. [PMID: 35955591 PMCID: PMC9369319 DOI: 10.3390/ijms23158458] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/19/2022] Open
Abstract
Carcinogenesis is a multistage process involving the dysregulation of multiple genes, proteins, and pathways that make any normal cell acquire a cancer cell phenotype. Therefore, it is no surprise that numerous ion channels could be involved in this process. Since their discovery and subsequent cloning, ion channels have been established as therapeutic targets in excitable cell pathologies (e.g., cardiac arrhythmias or epilepsy); however, their involvement in non-excitable cell pathologies is relatively recent. Among all ion channels, the voltage-gated potassium channels Kv10.1 have been established as a promising target in cancer treatment due to their high expression in tumoral tissues compared to low levels in healthy tissues.
Collapse
Affiliation(s)
- Enoch Luis
- Cátedras CONACYT—Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, C.U., Ciudad de México 04510, Mexico
- Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, C.U., Ciudad de México 04510, Mexico; (P.L.-S.); (M.L.D.-P.)
- Correspondence:
| | - Arely Anaya-Hernández
- Centro de Investigación en Genética y Ambiente, Universidad Autónoma de Tlaxcala, Km. 10.5 Autopista Tlaxcala-San Martín, Tlaxcala 90120, Mexico;
| | - Paulina León-Sánchez
- Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, C.U., Ciudad de México 04510, Mexico; (P.L.-S.); (M.L.D.-P.)
| | - María Luisa Durán-Pastén
- Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, C.U., Ciudad de México 04510, Mexico; (P.L.-S.); (M.L.D.-P.)
| |
Collapse
|
20
|
Audero MM, Prevarskaya N, Fiorio Pla A. Ca 2+ Signalling and Hypoxia/Acidic Tumour Microenvironment Interplay in Tumour Progression. Int J Mol Sci 2022; 23:7377. [PMID: 35806388 PMCID: PMC9266881 DOI: 10.3390/ijms23137377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/18/2023] Open
Abstract
Solid tumours are characterised by an altered microenvironment (TME) from the physicochemical point of view, displaying a highly hypoxic and acidic interstitial fluid. Hypoxia results from uncontrolled proliferation, aberrant vascularization and altered cancer cell metabolism. Tumour cellular apparatus adapts to hypoxia by altering its metabolism and behaviour, increasing its migratory and metastatic abilities by the acquisition of a mesenchymal phenotype and selection of aggressive tumour cell clones. Extracellular acidosis is considered a cancer hallmark, acting as a driver of cancer aggressiveness by promoting tumour metastasis and chemoresistance via the selection of more aggressive cell phenotypes, although the underlying mechanism is still not clear. In this context, Ca2+ channels represent good target candidates due to their ability to integrate signals from the TME. Ca2+ channels are pH and hypoxia sensors and alterations in Ca2+ homeostasis in cancer progression and vascularization have been extensively reported. In the present review, we present an up-to-date and critical view on Ca2+ permeable ion channels, with a major focus on TRPs, SOCs and PIEZO channels, which are modulated by tumour hypoxia and acidosis, as well as the consequent role of the altered Ca2+ signals on cancer progression hallmarks. We believe that a deeper comprehension of the Ca2+ signalling and acidic pH/hypoxia interplay will break new ground for the discovery of alternative and attractive therapeutic targets.
Collapse
Affiliation(s)
- Madelaine Magalì Audero
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Natalia Prevarskaya
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
| | - Alessandra Fiorio Pla
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| |
Collapse
|
21
|
Sudarikova AV, Bychkov ML, Kulbatskii DS, Chubinskiy-Nadezhdin VI, Shlepova OV, Shulepko MA, Koshelev SG, Kirpichnikov MP, Lyukmanova EN. Mambalgin-2 Inhibits Lung Adenocarcinoma Growth and Migration by Selective Interaction With ASIC1/α-ENaC/γ-ENaC Heterotrimer. Front Oncol 2022; 12:904742. [PMID: 35837090 PMCID: PMC9273970 DOI: 10.3389/fonc.2022.904742] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/24/2022] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is one of the most common cancer types in the world. Despite existing treatment strategies, overall patient survival remains low and new targeted therapies are required. Acidification of the tumor microenvironment drives the growth and metastasis of many cancers. Acid sensors such as acid-sensing ion channels (ASICs) may become promising targets for lung cancer therapy. Previously, we showed that inhibition of the ASIC1 channels by a recombinant analogue of mambalgin-2 from Dendroaspis polylepis controls oncogenic processes in leukemia, glioma, and melanoma cells. Here, we studied the effects and molecular targets of mambalgin-2 in lung adenocarcinoma A549 and Lewis cells, lung transformed WI-38 fibroblasts, and lung normal HLF fibroblasts. We found that mambalgin-2 inhibits the growth and migration of A549, metastatic Lewis P29 cells, and WI-38 cells, but not of normal fibroblasts. A549, Lewis, and WI-38 cells expressed different ASIC and ENaC subunits, while normal fibroblasts did not at all. Mambalgin-2 induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma cells. In line, acidification-evoked inward currents were observed only in A549 and WI-38 cells. Gene knockdown showed that the anti-proliferative and anti-migratory activity of mambalgin-2 is dependent on the expression of ASIC1a, α-ENaC, and γ-ENaC. Using affinity extraction and immunoprecipitation, mambalgin-2 targeting of ASIC1a/α-ENaC/γ-ENaC heteromeric channels in A549 cells was shown. Electrophysiology studies in Xenopus oocytes revealed that mambalgin-2 inhibits the ASIC1a/α-ENaC/γ-ENaC channels with higher efficacy than the ASIC1a channels, pointing on the heteromeric channels as a primary target of the toxin in cancer cells. Finally, bioinformatics analysis showed that the increased expression of ASIC1 and γ-ENaC correlates with a worse survival prognosis for patients with lung adenocarcinoma. Thus, the ASIC1a/α-ENaC/γ-ENaC heterotrimer can be considered a marker of cell oncogenicity and its targeting is promising for the design of new selective cancer therapeutics.
Collapse
Affiliation(s)
- Anastasia V. Sudarikova
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Group of Ionic Mechanisms of Cell Signaling, Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Maxim L. Bychkov
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Dmitrii S. Kulbatskii
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vladislav I. Chubinskiy-Nadezhdin
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Group of Ionic Mechanisms of Cell Signaling, Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga V. Shlepova
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
| | - Mikhail A. Shulepko
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sergey G. Koshelev
- Laboratory of Neuroreceptors and Neuroregulators, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Mikhail P. Kirpichnikov
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina N. Lyukmanova
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
- *Correspondence: Ekaterina N. Lyukmanova,
| |
Collapse
|
22
|
Ion Channel Drugs Suppress Cancer Phenotype in NG108-15 and U87 Cells: Toward Novel Electroceuticals for Glioblastoma. Cancers (Basel) 2022; 14:cancers14061499. [PMID: 35326650 PMCID: PMC8946312 DOI: 10.3390/cancers14061499] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma is a lethal brain cancer that commonly recurs after tumor resection and chemotherapy treatment. Depolarized resting membrane potentials and an acidic intertumoral extracellular pH have been associated with a proliferative state and drug resistance, suggesting that forced hyperpolarization and disruption of proton pumps in the plasma membrane could be a successful strategy for targeting glioblastoma overgrowth. We screened 47 compounds and compound combinations, most of which were ion-modulating, at different concentrations in the NG108-15 rodent neuroblastoma/glioma cell line. A subset of these were tested in the U87 human glioblastoma cell line. A FUCCI cell cycle reporter was stably integrated into both cell lines to monitor proliferation and cell cycle response. Immunocytochemistry, electrophysiology, and a panel of physiological dyes reporting voltage, calcium, and pH were used to characterize responses. The most effective treatments on proliferation in U87 cells were combinations of NS1643 and pantoprazole; retigabine and pantoprazole; and pantoprazole or NS1643 with temozolomide. Marker analysis and physiological dye signatures suggest that exposure to bioelectric drugs significantly reduces proliferation, makes the cells senescent, and promotes differentiation. These results, along with the observed low toxicity in human neurons, show the high efficacy of electroceuticals utilizing combinations of repurposed FDA approved drugs.
Collapse
|
23
|
Liao Y, Jing T, Zhang F, He P. In Situ Monitoring of Extracellular K + Using the Potentiometric Mode of Scanning Electrochemical Microscopy with a Carbon-Based Potassium Ion-Selective Tip. Anal Chem 2022; 94:4078-4086. [PMID: 35213803 DOI: 10.1021/acs.analchem.2c00002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The expression of potassium channels can be related to the occurrence and development of tumors. Their change would affect K+ outflow. Thus, in situ monitoring of extracellular K+ shows a great significance. Herein, the dual-functional K+ ion-selective electrode as the scanning electrochemical microscopy (SECM) tip (K+-ISE SECM tip) has been developed for in situ monitoring of the extracellular K+. Based on multi-wall carbon nanotubes as a transduction layer, the K+-ISE SECM tip realizes both the plotting of approach curves to position the tip for in situ detection and the recording of potential responses. It shows a near Nernstian response, good selectivity, and excellent stability. Based on these characteristics, it was used to in situ monitor K+ concentrations ([K+]o) of three breast cancer cell lines (MCF-7, MDA-MB-231, and SK-BR-3 cells) at 3 μm above the cell, and [K+]o of MDA-MB-231 cells show the highest value, followed by MCF-7 cells and SK-BR-3 cells. K+ outflow induced by electrical stimulation or pH changes of the culture environment (Δ[K+]o) was further determined, and the possible mechanism of K+ outflow was investigated with 4-aminopyridin (4-AP). MCF-7 cells present the largest value of Δ[K+]o, followed by MDA-MB-231 cells and SK-BR-3 cells at all the stimulation potentials, and pH 6.50 shows the greatest impact on K+ outflow of the three cell lines. The pretreatment of 4-AP changed K+ outflow, probably due to the regulation of voltage-gated channels. These findings provide insight into a deep understanding of the microenvironment influence on K+ outflow, thereby reflecting the possible mechanism of potassium channels.
Collapse
Affiliation(s)
- Yuxian Liao
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P.R. China
| | - Ting Jing
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P.R. China
| | - Fan Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P.R. China
| | - Pingang He
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P.R. China
| |
Collapse
|
24
|
Kittl M, Winklmayr M, Preishuber-Pflügl J, Strobl V, Gaisberger M, Ritter M, Jakab M. Low pH Attenuates Apoptosis by Suppressing the Volume-Sensitive Outwardly Rectifying (VSOR) Chloride Current in Chondrocytes. Front Cell Dev Biol 2022; 9:804105. [PMID: 35186954 PMCID: PMC8847443 DOI: 10.3389/fcell.2021.804105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/31/2021] [Indexed: 11/25/2022] Open
Abstract
In a variety of physiological and pathophysiological conditions, cells are exposed to acidic environments. Severe synovial fluid acidification also occurs in a progressive state of osteoarthritis (OA) affecting articular chondrocytes. In prior studies extracellular acidification has been shown to protect cells from apoptosis but the underlying mechanisms remain elusive. In the present study, we demonstrate that the inhibition of Cl− currents plays a significant role in the antiapoptotic effect of acidification in human articular chondrocytes. Drug-induced apoptosis was analyzed after exposure to staurosporine by caspase 3/7 activity and by annexin-V/7-actinomycin D (7-AAD) staining, followed by flow cytometry. Cell viability was assessed by resazurin, CellTiter-Glo and CellTiter-Fluor assays. Cl− currents and the mean cell volume were determined using the whole cell patch clamp technique and the Coulter method, respectively. The results reveal that in C28/I2 cells extracellular acidification decreases caspase 3/7 activity, enhances cell viability following staurosporine treatment and gradually deactivates the volume-sensitive outwardly rectifying (VSOR) Cl− current. Furthermore, the regulatory volume decrease (RVD) as well as the apoptotic volume decrease (ADV), which represents an early event during apoptosis, were absent under acidic conditions after hypotonicity-induced cell swelling and staurosporine-induced apoptosis, respectively. Like acidosis, the VSOR Cl− current inhibitor DIDS rescued chondrocytes from apoptotic cell death and suppressed AVD after induction of apoptosis with staurosporine. Similar to acidosis and DIDS, the VSOR channel blockers NPPB, niflumic acid (NFA) and DCPIB attenuated the staurosporine-induced AVD. NPPB and NFA also suppressed staurosporine-induced caspase 3/7 activation, while DCPIB and Tamoxifen showed cytotoxic effects per se. From these data, we conclude that the deactivation of VSOR Cl− currents impairs cell volume regulation under acidic conditions, which is likely to play an important role in the survivability of human articular chondrocytes.
Collapse
Affiliation(s)
- Michael Kittl
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
- *Correspondence: Michael Kittl,
| | - Martina Winklmayr
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
| | - Julia Preishuber-Pflügl
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Victoria Strobl
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Martin Gaisberger
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology, Pathophysiology and Biophysics—Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Martin Jakab
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology—Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Salzburg, Austria
| |
Collapse
|
25
|
Barbonari S, D'Amore A, Palombi F, De Cesaris P, Parrington J, Riccioli A, Filippini A. RELEVANCE OF LYSOSOMAL Ca2+ SIGNALLING MACHINERY IN CANCER. Cell Calcium 2022; 102:102539. [DOI: 10.1016/j.ceca.2022.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/23/2022]
|
26
|
Rychkov GY, Zhou FH, Adams MK, Brierley SM, Ma L, Barritt GJ. Orai1- and Orai2-, but not Orai3-mediated I CRAC is regulated by intracellular pH. J Physiol 2021; 600:623-643. [PMID: 34877682 DOI: 10.1113/jp282502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Three Orai (Orai1, Orai2, and Orai3) and two stromal interaction molecule (STIM1 and STIM2) mammalian protein homologues constitute major components of the store-operated Ca2+ entry mechanism. When co-expressed with STIM1, Orai1, Orai2 and Orai3 form highly selective Ca2+ channels with properties of Ca2+ release-activated Ca2+ (CRAC) channels. Despite the high level of homology between Orai proteins, CRAC channels formed by different Orai isoforms have distinctive properties, particularly with regards to Ca2+ -dependent inactivation, inhibition/potentiation by 2-aminoethyl diphenylborinate and sensitivity to reactive oxygen species. This study characterises and compares the regulation of Orai1, Orai2- and Orai3-mediated CRAC current (ICRAC ) by intracellular pH (pHi ). Using whole-cell patch clamping of HEK293T cells heterologously expressing Orai and STIM1, we show that ICRAC formed by each Orai homologue has a unique sensitivity to changes in pHi . Orai1-mediated ICRAC exhibits a strong dependence on pHi of both current amplitude and the kinetics of Ca2+ -dependent inactivation. In contrast, Orai2 amplitude, but not kinetics, depends on pHi , whereas Orai3 shows no dependence on pHi at all. Investigation of different Orai1-Orai3 chimeras suggests that pHi dependence of Orai1 resides in both the N-terminus and intracellular loop 2, and may also involve pH-dependent interactions with STIM1. KEY POINTS: It has been shown previously that Orai1/stromal interaction molecule 1 (STIM1)-mediated Ca2+ release-activated Ca2+ current (ICRAC ) is inhibited by intracellular acidification and potentiated by intracellular alkalinisation. The present study reveals that CRAC channels formed by each of the Orai homologues Orai1, Orai2 and Orai3 has a unique sensitivity to changes in intracellular pH (pHi ). The amplitude of Orai2 current is affected by the changes in pHi similarly to the amplitude of Orai1. However, unlike Orai1, fast Ca2+ -dependent inactivation of Orai2 is unaffected by acidic pHi . In contrast to both Orai1 and Orai2, Orai3 is not sensitive to pHi changes. Domain swapping between Orai1 and Orai3 identified the N-terminus and intracellular loop 2 as the molecular structures responsible for Orai1 regulation by pHi . Reduction of ICRAC dependence on pHi seen in a STIM1-independent Orai1 mutant suggested that some parts of STIM1 are also involved in ICRAC modulation by pHi .
Collapse
Affiliation(s)
- Grigori Y Rychkov
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Fiona H Zhou
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Melissa K Adams
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Stuart M Brierley
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
| | - Linlin Ma
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Greg J Barritt
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| |
Collapse
|
27
|
Panda S, Chatterjee O, Roy L, Chatterjee S. Targeting Ca 2+ signaling: A new arsenal against cancer. Drug Discov Today 2021; 27:923-934. [PMID: 34793973 DOI: 10.1016/j.drudis.2021.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/24/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
The drug resistance of cancer cells is a major concern in medical oncology, resulting in the failure of chemotherapy. Ca2+ plays a pivotal role in inducing multidrug resistance in cancer cells. Calcium signaling is a critical regulator of many cancer hallmarks, such as angiogenesis, invasiveness, and migration. In this review, we describe the involvement of Ca2+ signaling and associated proteins in cancer progression and in the development of multidrug resistance in cancer cells. We also highlight the possibilities and challenges of targeting the Ca2+ channels, transporters, and pumps involved in Ca2+ signaling in cancer cells through structure-based drug design. This work will open a new therapeutic window to be used against cancer in upcoming years.
Collapse
Affiliation(s)
- Suman Panda
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Oishika Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Laboni Roy
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Subhrangsu Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India.
| |
Collapse
|
28
|
Chokshi R, Bennett O, Zhelay T, Kozak JA. NSAIDs Naproxen, Ibuprofen, Salicylate, and Aspirin Inhibit TRPM7 Channels by Cytosolic Acidification. Front Physiol 2021; 12:727549. [PMID: 34733174 PMCID: PMC8558630 DOI: 10.3389/fphys.2021.727549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/10/2021] [Indexed: 01/23/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are used for relieving pain and inflammation accompanying numerous disease states. The primary therapeutic mechanism of these widely used drugs is the inhibition of cyclooxygenase 1 and 2 (COX1, 2) enzymes that catalyze the conversion of arachidonic acid into prostaglandins. At higher doses, NSAIDs are used for prevention of certain types of cancer and as experimental treatments for Alzheimer’s disease. In the immune system, various NSAIDs have been reported to influence neutrophil function and lymphocyte proliferation, and affect ion channels and cellular calcium homeostasis. Transient receptor potential melastatin 7 (TRPM7) cation channels are highly expressed in T lymphocytes and are inhibited by Mg2+, acidic pH, and polyamines. Here, we report a novel effect of naproxen, ibuprofen, salicylate, and acetylsalicylate on TRPM7. At concentrations of 3–30mM, they reversibly inhibited TRPM7 channel currents. By measuring intracellular pH with the ratiometric indicator BCECF, we found that at 300μM to 30mM, these NSAIDs reversibly acidified the cytoplasm in a concentration-dependent manner, and propose that TRPM7 channel inhibition is a consequence of cytosolic acidification, rather than direct. NSAID inhibition of TRPM7 channels was slow, voltage-independent, and displayed use-dependence, increasing in potency upon repeated drug applications. The extent of channel inhibition by salicylate strongly depended on cellular PI(4,5)P2 levels, as revealed when this phospholipid was depleted with voltage-sensitive lipid phosphatase (VSP). Salicylate inhibited heterologously expressed wildtype TRPM7 channels but not the S1107R variant, which is insensitive to cytosolic pH, Mg2+, and PI(4,5)P2 depletion. NSAID-induced acidification was also observed in Schneider 2 cells from Drosophila, an organism that lacks orthologous COX genes, suggesting that this effect is unrelated to COX enzyme activity. A 24-h exposure to 300μM–10mM naproxen resulted in a concentration-dependent reduction in cell viability. In addition to TRPM7, the described NSAID effect would be expected to apply to other ion channels and transporters sensitive to intracellular pH.
Collapse
Affiliation(s)
- Rikki Chokshi
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| | - Orville Bennett
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| | - Tetyana Zhelay
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| | - J Ashot Kozak
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| |
Collapse
|
29
|
Rodat-Despoix L, Chamlali M, Ouadid-Ahidouch H. Ion channels as key partners of cytoskeleton in cancer disease. Biochim Biophys Acta Rev Cancer 2021; 1876:188627. [PMID: 34520803 DOI: 10.1016/j.bbcan.2021.188627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
Several processes occur during tumor development including changes in cell morphology, a reorganization of the expression and distribution of the cytoskeleton proteins as well as ion channels. If cytoskeleton proteins and ion channels have been widely investigated in understanding cancer mechanisms, the interaction between these two elements and the identification of the associated signaling pathways are only beginning to emerge. In this review, we summarize the work published over the past 15 years relating to the roles played by ion channels in these mechanisms of reorganization of the cellular morphology, essential to metastatic dissemination, both through the physical interactions with elements of the cytoskeleton and by intracellular signaling pathways involved.
Collapse
Affiliation(s)
- Lise Rodat-Despoix
- Laboratoire de Physiologie Cellulaire et Moléculaire (UR 4667), Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France.
| | - Mohamed Chamlali
- Laboratoire de Physiologie Cellulaire et Moléculaire (UR 4667), Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
| | - Halima Ouadid-Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire (UR 4667), Université de Picardie Jules Verne, UFR des Sciences, 33 Rue St Leu, 80039 Amiens, France
| |
Collapse
|
30
|
Di Pompo G, Cortini M, Baldini N, Avnet S. Acid Microenvironment in Bone Sarcomas. Cancers (Basel) 2021; 13:cancers13153848. [PMID: 34359749 PMCID: PMC8345667 DOI: 10.3390/cancers13153848] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Although rare, malignant bone sarcomas have devastating clinical implications for the health and survival of young adults and children. To date, efforts to identify the molecular drivers and targets have focused on cancer cells or on the interplay between cancer cells and stromal cells in the tumour microenvironment. On the contrary, in the current literature, the role of the chemical-physical conditions of the tumour microenvironment that may be implicated in sarcoma aggressiveness and progression are poorly reported and discussed. Among these, extracellular acidosis is a well-recognized hallmark of bone sarcomas and promotes cancer growth and dissemination but data presented on this topic are fragmented. Hence, we intended to provide a general and comprehensive overview of the causes and implications of acidosis in bone sarcoma. Abstract In bone sarcomas, extracellular proton accumulation is an intrinsic driver of malignancy. Extracellular acidosis increases stemness, invasion, angiogenesis, metastasis, and resistance to therapy of cancer cells. It reprograms tumour-associated stroma into a protumour phenotype through the release of inflammatory cytokines. It affects bone homeostasis, as extracellular proton accumulation is perceived by acid-sensing ion channels located at the cell membrane of normal bone cells. In bone, acidosis results from the altered glycolytic metabolism of bone cancer cells and the resorption activity of tumour-induced osteoclasts that share the same ecosystem. Proton extrusion activity is mediated by extruders and transporters located at the cell membrane of normal and transformed cells, including vacuolar ATPase and carbonic anhydrase IX, or by the release of highly acidic lysosomes by exocytosis. To date, a number of investigations have focused on the effects of acidosis and its inhibition in bone sarcomas, including studies evaluating the use of photodynamic therapy. In this review, we will discuss the current status of all findings on extracellular acidosis in bone sarcomas, with a specific focus on the characteristics of the bone microenvironment and the acid-targeting therapeutic approaches that are currently being evaluated.
Collapse
Affiliation(s)
- Gemma Di Pompo
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Margherita Cortini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Nicola Baldini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
- Correspondence:
| |
Collapse
|
31
|
Hofschröer V, Najder K, Rugi M, Bouazzi R, Cozzolino M, Arcangeli A, Panyi G, Schwab A. Ion Channels Orchestrate Pancreatic Ductal Adenocarcinoma Progression and Therapy. Front Pharmacol 2021; 11:586599. [PMID: 33841132 PMCID: PMC8025202 DOI: 10.3389/fphar.2020.586599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a dismal prognosis. Therapeutic interventions are largely ineffective. A better understanding of the pathophysiology is required. Ion channels contribute substantially to the "hallmarks of cancer." Their expression is dysregulated in cancer, and they are "misused" to drive cancer progression, but the underlying mechanisms are unclear. Ion channels are located in the cell membrane at the interface between the intracellular and extracellular space. They sense and modify the tumor microenvironment which in itself is a driver of PDAC aggressiveness. Ion channels detect, for example, locally altered proton and electrolyte concentrations or mechanical stimuli and transduce signals triggered by these microenvironmental cues through association with intracellular signaling cascades. While these concepts have been firmly established for other cancers, evidence has emerged only recently that ion channels are drivers of PDAC aggressiveness. Particularly, they appear to contribute to two of the characteristic PDAC features: the massive fibrosis of the tumor stroma (desmoplasia) and the efficient immune evasion. Our critical review of the literature clearly shows that there is still a remarkable lack of knowledge with respect to the contribution of ion channels to these two typical PDAC properties. Yet, we can draw parallels from ion channel research in other fibrotic and inflammatory diseases. Evidence is accumulating that pancreatic stellate cells express the same "profibrotic" ion channels. Similarly, it is at least in part known which major ion channels are expressed in those innate and adaptive immune cells that populate the PDAC microenvironment. We explore potential therapeutic avenues derived thereof. Since drugs targeting PDAC-relevant ion channels are already in clinical use, we propose to repurpose those in PDAC. The quest for ion channel targets is both motivated and complicated by the fact that some of the relevant channels, for example, KCa3.1, are functionally expressed in the cancer, stroma, and immune cells. Only in vivo studies will reveal which arm of the balance we should put our weights on when developing channel-targeting PDAC therapies. The time is up to explore the efficacy of ion channel targeting in (transgenic) murine PDAC models before launching clinical trials with repurposed drugs.
Collapse
Affiliation(s)
| | - Karolina Najder
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Micol Rugi
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Rayhana Bouazzi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Florence, Italy
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
32
|
Alvarez-Arellano L, Salazar-García M, Corona JC. Neuroprotective Effects of Quercetin in Pediatric Neurological Diseases. Molecules 2020; 25:E5597. [PMID: 33260783 PMCID: PMC7731313 DOI: 10.3390/molecules25235597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is a crucial event underlying several pediatric neurological diseases, such as the central nervous system (CNS) tumors, autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD). Neuroprotective therapy with natural compounds used as antioxidants has the potential to delay, ameliorate or prevent several pediatric neurological diseases. The present review provides an overview of the most recent research outcomes following quercetin treatment for CNS tumors, ASD and ADHD as well as describes the potential in vitro and in vivo ameliorative effect on oxidative stress of bioactive natural compounds, which seems like a promising future therapy for these diseases. The neuroprotective effects of quercetin against oxidative stress can also be applied in the management of several neurodegenerative disorders with effects such as anti-cancer, anti-inflammatory, anti-viral, anti-obesity and anti-microbial. Therefore, quercetin appears to be a suitable adjuvant for therapy against pediatric neurological diseases.
Collapse
Affiliation(s)
| | - Marcela Salazar-García
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
33
|
Harguindey S, Alfarouk K, Polo Orozco J, Fais S, Devesa J. Towards an Integral Therapeutic Protocol for Breast Cancer Based upon the New H +-Centered Anticancer Paradigm of the Late Post-Warburg Era. Int J Mol Sci 2020; 21:E7475. [PMID: 33050492 PMCID: PMC7589677 DOI: 10.3390/ijms21207475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
A brand new approach to the understanding of breast cancer (BC) is urgently needed. In this contribution, the etiology, pathogenesis, and treatment of this disease is approached from the new pH-centric anticancer paradigm. Only this unitarian perspective, based upon the hydrogen ion (H+) dynamics of cancer, allows for the understanding and integration of the many dualisms, confusions, and paradoxes of the disease. The new H+-related, wide-ranging model can embrace, from a unique perspective, the many aspects of the disease and, at the same time, therapeutically interfere with most, if not all, of the hallmarks of cancer known to date. The pH-related armamentarium available for the treatment of BC reviewed here may be beneficial for all types and stages of the disease. In this vein, we have attempted a megasynthesis of traditional and new knowledge in the different areas of breast cancer research and treatment based upon the wide-ranging approach afforded by the hydrogen ion dynamics of cancer. The concerted utilization of the pH-related drugs that are available nowadays for the treatment of breast cancer is advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Department of Pharmacology, Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|