1
|
Blažič A, Guinard M, Leskovar T, O'Connor RP, Rems L. Long-term changes in transmembrane voltage after electroporation are governed by the interplay between nonselective leak current and ion channel activation. Bioelectrochemistry 2025; 161:108802. [PMID: 39243733 DOI: 10.1016/j.bioelechem.2024.108802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Electroporation causes a temporal increase in cell membrane permeability and leads to prolonged changes in transmembrane voltage (TMV) in both excitable and non-excitable cells. However, the mechanisms of these TMV changes remain to be fully elucidated. To this end, we monitored TMV over 30 min after exposing two different cell lines to a single 100 µs electroporation pulse using the FLIPR Membrane Potential dye. In CHO-K1 cells, which express very low levels of endogenous ion channels, membrane depolarization following pulse exposure could be explained by nonselective leak current, which persists until the membrane reseals, enabling the cells to recover their resting TMV. In U-87 MG cells, which express many different ion channels, we unexpectedly observed membrane hyperpolarization following the initial depolarization phase, but only at 33 °C and not at 25 °C. We developed a theoretical model, supported by experiments with ion channel inhibitors, which indicated that hyperpolarization could largely be attributed to the activation of calcium-activated potassium channels. Ion channel activation, coupled with changes in TMV and intracellular calcium, participates in various physiological processes, including cell proliferation, differentiation, migration, and apoptosis. Therefore, our study suggests that ion channels could present a potential target for influencing the biological response after electroporation.
Collapse
Affiliation(s)
- Anja Blažič
- University of Ljubljana, Faculty of Electrical Engineering, SI-1000 Ljubljana, Slovenia
| | - Manon Guinard
- University of Ljubljana, Faculty of Electrical Engineering, SI-1000 Ljubljana, Slovenia
| | - Tomaž Leskovar
- University of Ljubljana, Faculty of Electrical Engineering, SI-1000 Ljubljana, Slovenia
| | - Rodney P O'Connor
- Mines Saint-Etienne, Centre CMP, Département BEL, F-13541 Gardanne, France
| | - Lea Rems
- University of Ljubljana, Faculty of Electrical Engineering, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
2
|
Michelucci A, Catacuzzeno L. Piezo1, the new actor in cell volume regulation. Pflugers Arch 2024; 476:1023-1039. [PMID: 38581527 PMCID: PMC11166825 DOI: 10.1007/s00424-024-02951-y] [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: 01/11/2024] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024]
Abstract
All animal cells control their volume through a complex set of mechanisms, both to counteract osmotic perturbations of the environment and to enable numerous vital biological processes, such as proliferation, apoptosis, and migration. The ability of cells to adjust their volume depends on the activity of ion channels and transporters which, by moving K+, Na+, and Cl- ions across the plasma membrane, generate the osmotic gradient that drives water in and out of the cell. In 2010, Patapoutian's group identified a small family of evolutionarily conserved, Ca2+-permeable mechanosensitive channels, Piezo1 and Piezo2, as essential components of the mechanically activated current that mediates mechanotransduction in vertebrates. Piezo1 is expressed in several tissues and its opening is promoted by a wide range of mechanical stimuli, including membrane stretch/deformation and osmotic stress. Piezo1-mediated Ca2+ influx is used by the cell to convert mechanical forces into cytosolic Ca2+ signals that control diverse cellular functions such as migration and cell death, both dependent on changes in cell volume and shape. The crucial role of Piezo1 in the regulation of cell volume was first demonstrated in erythrocytes, which need to reduce their volume to pass through narrow capillaries. In HEK293 cells, increased expression of Piezo1 was found to enhance the regulatory volume decrease (RVD), the process whereby the cell re-establishes its original volume after osmotic shock-induced swelling, and it does so through Ca2+-dependent modulation of the volume-regulated anion channels. More recently we reported that Piezo1 controls the RVD in glioblastoma cells via the modulation of Ca2+-activated K+ channels. To date, however, the mechanisms through which this mechanosensitive channel controls cell volume and maintains its homeostasis have been poorly investigated and are still far from being understood. The present review aims to provide a broad overview of the literature discussing the recent advances on this topic.
Collapse
Affiliation(s)
- A Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - L Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| |
Collapse
|
3
|
Van NTH, Kim WK, Nam JH. Challenges in the Therapeutic Targeting of KCa Channels: From Basic Physiology to Clinical Applications. Int J Mol Sci 2024; 25:2965. [PMID: 38474212 PMCID: PMC10932353 DOI: 10.3390/ijms25052965] [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: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/14/2024] Open
Abstract
Calcium-activated potassium (KCa) channels are ubiquitously expressed throughout the body and are able to regulate membrane potential and intracellular calcium concentrations, thereby playing key roles in cellular physiology and signal transmission. Consequently, it is unsurprising that KCa channels have been implicated in various diseases, making them potential targets for pharmaceutical interventions. Over the past two decades, numerous studies have been conducted to develop KCa channel-targeting drugs, including those for disorders of the central and peripheral nervous, cardiovascular, and urinary systems and for cancer. In this review, we synthesize recent findings regarding the structure and activating mechanisms of KCa channels. We also discuss the role of KCa channel modulators in therapeutic medicine. Finally, we identify the major reasons behind the delay in bringing these modulators to the pharmaceutical market and propose new strategies to promote their application.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| |
Collapse
|
4
|
Guerriero C, Fanfarillo R, Mancini P, Sterbini V, Guarguaglini G, Sforna L, Michelucci A, Catacuzzeno L, Tata AM. M2 muscarinic receptors negatively modulate cell migration in human glioblastoma cells. Neurochem Int 2024; 174:105673. [PMID: 38185384 DOI: 10.1016/j.neuint.2023.105673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/17/2023] [Accepted: 12/29/2023] [Indexed: 01/09/2024]
Abstract
Glioblastoma (GB) is a very aggressive human brain tumor. The high growth potential and invasiveness make this tumor surgically and pharmacologically untreatable. Our previous work demonstrated that the activation of the M2 muscarinic acetylcholine receptors (M2 mAChRs) inhibited cell proliferation and survival in GB cell lines and in the cancer stem cells derived from human biopsies. The aim of the present study was to investigate the ability of M2 mAChR to modulate cell migration in two different GB cell lines: U87 and U251. By wound healing assay and single cell migration analysis performed by time-lapse microscopy, we demonstrated the ability of M2 mAChRs to negatively modulate cell migration in U251 but not in the U87 cell line. In order to explain the different effects observed in the two cell lines we have evaluated the possible involvement of the intermediate conductance calcium-activated potassium (IKCa) channel. IKCa channel is present in the GB cells, and it has been demonstrated to modulate cell migration. Using the perforated patch-clamp technique we have found that selective activation of M2 mAChR significantly reduced functional density of the IKCa current in U251 but not in U87 cells. To understand whether the M2 mAChR mediated reduction of ion channel density in the U251 cell line was relevant for the cell migration impairment, we tested the effects of TRAM-34, a selective inhibitor of the IKCa channel, in wound healing assay. We found that it was able to markedly reduce U251 cell migration and significantly decrease the number of invadopodia-like structure formations. These results suggest that only in U251 cells the reduced cell migration M2 mAChR-mediated might involve, at least in part, the IKCa channel.
Collapse
Affiliation(s)
- Claudia Guerriero
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy.
| | - Rachele Fanfarillo
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy.
| | - Patrizia Mancini
- Department Experimental Medicine, Sapienza University of Rome, 00185, Rome, Italy.
| | | | | | - Luigi Sforna
- Department of Chemistry Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - Antonio Michelucci
- Department of Chemistry Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - Luigi Catacuzzeno
- Department of Chemistry Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy; Research Centre of Neurobiology Daniel Bovet, Sapienza University of Rome, 00185, Rome, Italy.
| |
Collapse
|
5
|
Stransky N, Ganser K, Quintanilla-Martinez L, Gonzalez-Menendez I, Naumann U, Eckert F, Koch P, Huber SM, Ruth P. Efficacy of combined tumor irradiation and K Ca3.1-targeting with TRAM-34 in a syngeneic glioma mouse model. Sci Rep 2023; 13:20604. [PMID: 37996600 PMCID: PMC10667541 DOI: 10.1038/s41598-023-47552-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
The intermediate-conductance calcium-activated potassium channel KCa3.1 has been proposed to be a new potential target for glioblastoma treatment. This study analyzed the effect of combined irradiation and KCa3.1-targeting with TRAM-34 in the syngeneic, immune-competent orthotopic SMA-560/VM/Dk glioma mouse model. Whereas neither irradiation nor TRAM-34 treatment alone meaningfully prolonged the survival of the animals, the combination significantly prolonged the survival of the mice. We found an irradiation-induced hyperinvasion of glioma cells into the brain, which was inhibited by concomitant TRAM-34 treatment. Interestingly, TRAM-34 did neither radiosensitize nor impair SMA-560's intrinsic migratory capacities in vitro. Exploratory findings hint at increased TGF-β1 signaling after irradiation. On top, we found a marginal upregulation of MMP9 mRNA, which was inhibited by TRAM-34. Last, infiltration of CD3+, CD8+ or FoxP3+ T cells was not impacted by either irradiation or KCa3.1 targeting and we found no evidence of adverse events of the combined treatment. We conclude that concomitant irradiation and TRAM-34 treatment is efficacious in this preclinical glioma model.
Collapse
Affiliation(s)
- Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, 72076, Tübingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Ulrike Naumann
- Molecular Neurooncology, Hertie Institute for Clinical Brain Research and Center Neurology, University of Tübingen, 72076, Tübingen, Germany
- Faculty of Medicine University, Gene and RNA Therapy Center (GRTC), Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
- Department of Radiation Oncology, Comprehensive Cancer Center, Medical University Vienna, AKH, Wien, Austria
| | - Pierre Koch
- Department of Pharmaceutical/Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
6
|
Younes S, Mourad N, Salla M, Rahal M, Hammoudi Halat D. Potassium Ion Channels in Glioma: From Basic Knowledge into Therapeutic Applications. MEMBRANES 2023; 13:434. [PMID: 37103862 PMCID: PMC10144598 DOI: 10.3390/membranes13040434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 06/19/2023]
Abstract
Ion channels, specifically those controlling the flux of potassium across cell membranes, have recently been shown to exhibit an important role in the pathophysiology of glioma, the most common primary central nervous system tumor with a poor prognosis. Potassium channels are grouped into four subfamilies differing by their domain structure, gating mechanisms, and functions. Pertinent literature indicates the vital functions of potassium channels in many aspects of glioma carcinogenesis, including proliferation, migration, and apoptosis. The dysfunction of potassium channels can result in pro-proliferative signals that are highly related to calcium signaling as well. Moreover, this dysfunction can feed into migration and metastasis, most likely by increasing the osmotic pressure of cells allowing the cells to initiate the "escape" and "invasion" of capillaries. Reducing the expression or channel blockage has shown efficacy in reducing the proliferation and infiltration of glioma cells as well as inducing apoptosis, priming several approaches to target potassium channels in gliomas pharmacologically. This review summarizes the current knowledge on potassium channels, their contribution to oncogenic transformations in glioma, and the existing perspectives on utilizing them as potential targets for therapy.
Collapse
Affiliation(s)
- Samar Younes
- Department of Biomedical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut 1103, Lebanon;
| | - Nisreen Mourad
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut 1103, Lebanon;
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
| | - Mohamed Salla
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Bekaa 146404, Lebanon;
| | - Mohamad Rahal
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
| | - Dalal Hammoudi Halat
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
- Academic Quality Department, QU Health, Qatar University, Doha 2713, Qatar;
| |
Collapse
|
7
|
Aquaporins and Ion Channels as Dual Targets in the Design of Novel Glioblastoma Therapeutics to Limit Invasiveness. Cancers (Basel) 2023; 15:cancers15030849. [PMID: 36765806 PMCID: PMC9913334 DOI: 10.3390/cancers15030849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Current therapies for Glioblastoma multiforme (GBM) focus on eradicating primary tumors using radiotherapy, chemotherapy and surgical resection, but have limited success in controlling the invasive spread of glioma cells into a healthy brain, the major factor driving short survival times for patients post-diagnosis. Transcriptomic analyses of GBM biopsies reveal clusters of membrane signaling proteins that in combination serve as robust prognostic indicators, including aquaporins and ion channels, which are upregulated in GBM and implicated in enhanced glioblastoma motility. Accumulating evidence supports our proposal that the concurrent pharmacological targeting of selected subclasses of aquaporins and ion channels could impede glioblastoma invasiveness by impairing key cellular motility pathways. Optimal sets of channels to be selected as targets for combined therapies could be tailored to the GBM cancer subtype, taking advantage of differences in patterns of expression between channels that are characteristic of GBM subtypes, as well as distinguishing them from non-cancerous brain cells such as neurons and glia. Focusing agents on a unique channel fingerprint in GBM would further allow combined agents to be administered at near threshold doses, potentially reducing off-target toxicity. Adjunct therapies which confine GBM tumors to their primary sites during clinical treatments would offer profound advantages for treatment efficacy.
Collapse
|
8
|
Massenzio F, Cambiaghi M, Marchiotto F, Boriero D, Limatola C, D’Alessandro G, Buffelli M. In vivo morphological alterations of TAMs during KCa3.1 inhibition-by using in vivo two-photon time-lapse technology. Front Cell Neurosci 2022; 16:1002487. [PMID: 36589283 PMCID: PMC9798303 DOI: 10.3389/fncel.2022.1002487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor associated macrophages (TAMs) are the mostprevalent cells recruited in the tumor microenvironment (TME). Once recruited, TAMs acquire a pro-tumor phenotype characterized by a typical morphology: ameboid in the tumor core and with larger soma and thick branches in the tumor periphery. Targeting TAMs by reverting them to an anti-tumor phenotype is a promising strategy for cancer immunotherapy. Taking advantage of Cx3cr1GFP/WT heterozygous mice implanted with murine glioma GL261-RFP cells we investigated the role of Ca2+-activated K+ channel (KCa3.1) on the phenotypic shift of TAMs at the late stage of glioma growth through in vivo two-photon imaging. We demonstrated that TAMs respond promptly to KCa3.1 inhibition using a selective inhibitor of the channel (TRAM-34) in a time-dependent manner by boosting ramified projections attributable to a less hypertrophic phenotype in the tumor core. We also revealed a selective effect of drug treatment by reducing both glioma cells and TAMs in the tumor core with no interference with surrounding cells. Taken together, our data indicate a TRAM-34-dependent progressive morphological transformation of TAMs toward a ramified and anti-tumor phenotype, suggesting that the timing of KCa3.1 inhibition is a key point to allow beneficial effects on TAMs.
Collapse
Affiliation(s)
- Francesca Massenzio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy,*Correspondence: Mario Buffelli Francesca Massenzio
| | - Marco Cambiaghi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federica Marchiotto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Diana Boriero
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy,IRCCS Neuromed, Pozzilli, Italy
| | - Giuseppina D’Alessandro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy,IRCCS Neuromed, Pozzilli, Italy
| | - Mario Buffelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy,*Correspondence: Mario Buffelli Francesca Massenzio
| |
Collapse
|
9
|
Yan P, Ke B, Fang X. Ion channels as a therapeutic target for renal fibrosis. Front Physiol 2022; 13:1019028. [PMID: 36277193 PMCID: PMC9581181 DOI: 10.3389/fphys.2022.1019028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Renal ion channel transport and electrolyte disturbances play an important role in the process of functional impairment and fibrosis in the kidney. It is well known that there are limited effective drugs for the treatment of renal fibrosis, and since a large number of ion channels are involved in the renal fibrosis process, understanding the mechanisms of ion channel transport and the complex network of signaling cascades between them is essential to identify potential therapeutic approaches to slow down renal fibrosis. This review summarizes the current work of ion channels in renal fibrosis. We pay close attention to the effect of cystic fibrosis transmembrane conductance regulator (CFTR), transmembrane Member 16A (TMEM16A) and other Cl− channel mediated signaling pathways and ion concentrations on fibrosis, as well as the various complex mechanisms for the action of Ca2+ handling channels including Ca2+-release-activated Ca2+ channel (CRAC), purinergic receptor, and transient receptor potential (TRP) channels. Furthermore, we also focus on the contribution of Na+ transport such as epithelial sodium channel (ENaC), Na+, K+-ATPase, Na+-H+ exchangers, and K+ channels like Ca2+-activated K+ channels, voltage-dependent K+ channel, ATP-sensitive K+ channels on renal fibrosis. Proposed potential therapeutic approaches through further dissection of these mechanisms may provide new therapeutic opportunities to reduce the burden of chronic kidney disease.
Collapse
|
10
|
Caglioti C, Palazzetti F, Monarca L, Lobello R, Ceccarini MR, Iannitti RG, Russo R, Ragonese F, Pennetta C, De Luca A, Codini M, Fioretti B. LY294002 Inhibits Intermediate Conductance Calcium-Activated Potassium (KCa3.1) Current in Human Glioblastoma Cells. Front Physiol 2022; 12:790922. [PMID: 35069252 PMCID: PMC8782274 DOI: 10.3389/fphys.2021.790922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Glioblastomas (GBs) are among the most common tumors with high malignancy and invasiveness of the central nervous system. Several alterations in protein kinase and ion channel activity are involved to maintain the malignancy. Among them, phosphatidylinositol 3-kinase (PI3K) activity and intermediate conductance calcium-activated potassium (KCa3.1) current are involved in several aspects of GB biology. By using the electrophysiological approach and noise analysis, we observed that KCa3.1 channel activity is LY294002-sensitive and Wortmannin-resistant in accordance with the involvement of PI3K class IIβ (PI3KC2β). This modulation was observed also during the endogenous activation of KCa3.1 current with histamine. The principal action of PI3KC2β regulation was the reduction of open probability in intracellular free calcium saturating concentration. An explanation based on the “three-gate” model of the KCa3.1 channel by PI3KC2β was proposed. Based on the roles of KCa3.1 and PI3KC2β in GB biology, a therapeutic implication was suggested to prevent chemo- and radioresistance mechanisms.
Collapse
Affiliation(s)
- Concetta Caglioti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.,Department of Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Federico Palazzetti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.,Department of Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | | | | | | | - Roberta Russo
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Chiara Pennetta
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Antonella De Luca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| |
Collapse
|
11
|
Sforna L, Michelucci A, Morena F, Argentati C, Franciolini F, Vassalli M, Martino S, Catacuzzeno L. Piezo1 controls cell volume and migration by modulating swelling-activated chloride current through Ca 2+ influx. J Cell Physiol 2021; 237:1857-1870. [PMID: 34913176 DOI: 10.1002/jcp.30656] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/21/2022]
Abstract
Regulatory volume decrease (RVD), a homeostatic process responsible for the re-establishment of the original cell volume upon swelling, is critical in controlling several functions, including migration. RVD is mainly sustained by the swelling-activated Cl- current (ICl,swell ), which can be modulated by cytoplasmic Ca2+ . Cell swelling also activates mechanosensitive channels, including the ubiquitously expressed Ca2+ -permeable channel Piezo1. We hypothesized that, by controlling cytoplasmic Ca2+ and in turn ICl,swell , Piezo1 is involved in the fine regulation of RVD and cell migration. We compared RVD and ICl,swell in wild-type (WT) HEK293T cells, which express endogenous levels of Piezo1, and in cells overexpressing (OVER) or knockout (KO) for Piezo1. Compared to WT, RVD was markedly increased in OVER, while virtually absent in KO cells. Consistently, ICl,swell amplitude was highest in OVER and lowest in KO cells, with WT cells displaying an intermediate level, suggesting a Ca2+ -dependent modulation of the current by Piezo1 channels. Indeed, in the absence of external Ca2+ , ICl,swell in both WT and OVER cells, as well as the RVD probed in OVER cells, were significantly lower than in the presence of Ca2+ and no longer different compared to KO cells. However, the Piezo-mediated Ca2+ influx was ineffective in enhancing ICl,swell in the absence of releasable Ca2+ from intracellular stores. The different expression levels of Piezo1 affected also cell migration which was strongly enhanced in OVER, while reduced in KO cells, as compared to WT. Taken together, our data indicate that Piezo1 controls RVD and migration in HEK293T cells by modulating ICl,swell through Ca2+ influx.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Antonio Michelucci
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti, Chieti, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Center for the Cellular Microenvironment, School of Engineering, G12 8LT, Glasgow, UK
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy.,CEMIN, Center of Excellence on Nanostructured Innovative Materials, University of Perugia, Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
12
|
Abstract
Neoplastic transformation is reportedly associated with alterations of the potassium transport across plasma and intracellular membranes. These alterations have been identified as crucial elements of the tumourigenic reprogramming of cells. Potassium channels may contribute to cancer initiation, malignant progression and therapy resistance of tumour cells. The book chapter focusses on (oncogenic) potassium channels frequently upregulated in different tumour entities, upstream and downstream signalling of these channels, their contribution to the maintenance of cancer stemness and the formation of an immunosuppressive tumour microenvironment. In addition, their role in adaptation to tumour hypoxia, metabolic reprogramming, as well as tumour spreading and metastasis is discussed. Finally, we discuss how (oncogenic) potassium channels may confer treatment resistance of tumours against radiation and chemotherapy and thus might be harnessed for new therapy strategies, for instance, by repurposing approved drugs known to target potassium channels.
Collapse
|
13
|
Chen X, Wang L, Cao L, Li T, Li Z, Sun Y, Ding J, Zhou C, Xie Y, Yue N, Nan J, Jia XM, Peng C, Li H, Yang J, Xiao H. Regulation of Anion Channel LRRC8 Volume-Regulated Anion Channels in Transport of 2'3'-Cyclic GMP-AMP and Cisplatin under Steady State and Inflammation. THE JOURNAL OF IMMUNOLOGY 2021; 206:2061-2074. [PMID: 33827893 DOI: 10.4049/jimmunol.2000989] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/18/2021] [Indexed: 11/19/2022]
Abstract
The recently identified anion channel LRRC8 volume-regulated anion channels (VRACs) are heteromeric hexamers constituted with the obligate LRRC8A subunit paired with at least one of the accessory LRRC8B to LRRC8E subunits. In addition to transport chloride, taurine, and glutamate, LRRC8 VRACs also transport the anticancer agent cisplatin and STING agonists 2'3'-cyclic GMP-AMP (cGAMP) and cyclic dinucleotides; hence, they are implicated in a variety of physiological and pathological processes, such as cell swelling, stroke, cancer, and viral infection. Although the subunit composition largely determines VRAC substrate specificity, the opening of various VRAC pores under physiological and pathological settings remains enigmatic. In this study, we demonstrated that VRACs comprising LRRC8A and LRRC8E (LRRC8A/E-containing VRACs), specialized in cGAMP transport, can be opened by a protein component present in serum under resting condition. Serum depletion ablated the tonic activity of LRRC8A/E-containing VRACs, decreasing cGAMP transport in various human and murine cells. Also, heating or proteinase K treatment abolished the ability of serum to activate VRAC. Genetic analyses revealed a crucial role for cGAMP synthase (cGAS) in serum/TNF-promoted VRAC activation. Notably, the presence of cGAS on the plasma membrane, rather than its DNA-binding or enzymatic activity, enabled VRAC activation. Moreover, phospholipid PIP2 seemed to be instrumental in the membrane localization of cGAS and its association with VRACs. Corroborating a role for LRRC8A/D-containing VRACs in cisplatin transport, serum and TNF markedly potentiated cisplatin uptake and killing of cancer cells derived from human or mouse. Together, these observations provide new insights into the complex regulation of VRAC activation and suggest a novel approach to enhance the efficacy of cGAMP and cisplatin in treating infection and cancer.
Collapse
Affiliation(s)
- Xia Chen
- Institute of Cancer Biology and Drug Screening, College of Life Sciences, Lanzhou University, Lanzhou, Gansu, China.,The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Li Wang
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Limin Cao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tiantian Li
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhihong Li
- Zhangjiang Lab, National Facility for Protein Science in Shanghai, Shanghai, China.,Shanghai Science Research Center, Chinese Academy of Sciences, Shanghai, China
| | - Yumeng Sun
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jinqiu Ding
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chun Zhou
- Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China; and
| | - Yadong Xie
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China; and
| | - Nan Yue
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Nan
- Institute of Cancer Biology and Drug Screening, College of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xin-Ming Jia
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chao Peng
- Zhangjiang Lab, National Facility for Protein Science in Shanghai, Shanghai, China.,Shanghai Science Research Center, Chinese Academy of Sciences, Shanghai, China
| | - Huabin Li
- Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China; and
| | - Jinbo Yang
- Institute of Cancer Biology and Drug Screening, College of Life Sciences, Lanzhou University, Lanzhou, Gansu, China; .,Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Hui Xiao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China;
| |
Collapse
|
14
|
Alharbi A, Zhang Y, Parrington J. Deciphering the Role of Ca 2+ Signalling in Cancer Metastasis: From the Bench to the Bedside. Cancers (Basel) 2021; 13:E179. [PMID: 33430230 PMCID: PMC7825727 DOI: 10.3390/cancers13020179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 01/03/2023] Open
Abstract
Metastatic cancer is one of the major causes of cancer-related mortalities. Metastasis is a complex, multi-process phenomenon, and a hallmark of cancer. Calcium (Ca2+) is a ubiquitous secondary messenger, and it has become evident that Ca2+ signalling plays a vital role in cancer. Ca2+ homeostasis is dysregulated in physiological processes related to tumour metastasis and progression-including cellular adhesion, epithelial-mesenchymal transition, cell migration, motility, and invasion. In this review, we looked at the role of intracellular and extracellular Ca2+ signalling pathways in processes that contribute to metastasis at the local level and also their effects on cancer metastasis globally, as well as at underlying molecular mechanisms and clinical applications. Spatiotemporal Ca2+ homeostasis, in terms of oscillations or waves, is crucial for hindering tumour progression and metastasis. They are a limited number of clinical trials investigating treating patients with advanced stages of various cancer types. Ca2+ signalling may serve as a novel hallmark of cancer due to the versatility of Ca2+ signals in cells, which suggests that the modulation of specific upstream/downstream targets may be a therapeutic approach to treat cancer, particularly in patients with metastatic cancers.
Collapse
Affiliation(s)
- Abeer Alharbi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
- Pharmaceutical Sciences Department, College of Pharmacy, King Saud Bin Abdul-Aziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Yuxuan Zhang
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| |
Collapse
|
15
|
Brandalise F, Ratto D, Leone R, Olivero F, Roda E, Locatelli CA, Grazia Bottone M, Rossi P. Deeper and Deeper on the Role of BK and Kir4.1 Channels in Glioblastoma Invasiveness: A Novel Summative Mechanism? Front Neurosci 2020; 14:595664. [PMID: 33328867 PMCID: PMC7734145 DOI: 10.3389/fnins.2020.595664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
In the last decades, increasing evidence has revealed that a large number of channel protein and ion pumps exhibit impaired expression in cancers. This dysregulation is responsible for high proliferative rates as well as migration and invasiveness, reflected in the recently coined term oncochannelopathies. In glioblastoma (GBM), the most invasive and aggressive primary brain tumor, GBM cells modify their ionic equilibrium in order to change their volume as a necessary step prior to migration. This mechanism involves increased expression of BK channels and downregulation of the normally widespread Kir4.1 channels, as noted in GBM biopsies from patients. Despite a large body of work implicating BK channels in migration in response to an artificial intracellular calcium rise, little is known about how this channel acts in GBM cells at resting membrane potential (RMP), as compared to other channels that are constitutively open, such as Kir4.1. In this review we propose that a residual fraction of functionally active Kir4.1 channels mediates a small, but continuous, efflux of potassium at the more depolarized RMP of GBM cells. In addition, coinciding with transient membrane deformation and the intracellular rise in calcium concentration, brief activity of BK channels can induce massive and rapid cytosolic water loss that reduces cell volume (cell shrinkage), a necessary step for migration within the brain parenchyma.
Collapse
Affiliation(s)
- Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Daniela Ratto
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| | - Roberta Leone
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Federico Olivero
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy.,Pavia Poison Centre, National Toxicology Information Centre, Laboratory of Clinical & Experimental Toxicology, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Alessandro Locatelli
- Pavia Poison Centre, National Toxicology Information Centre, Laboratory of Clinical & Experimental Toxicology, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| |
Collapse
|
16
|
Catacuzzeno L, Sforna L, Esposito V, Limatola C, Franciolini F. Ion Channels in Glioma Malignancy. Rev Physiol Biochem Pharmacol 2020; 181:223-267. [DOI: 10.1007/112_2020_44] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Ratto D, Ferrari B, Roda E, Brandalise F, Siciliani S, De Luca F, Priori EC, Di Iorio C, Cobelli F, Veneroni P, Bottone MG, Rossi P. Squaring the Circle: A New Study of Inward and Outward-Rectifying Potassium Currents in U251 GBM Cells. Cell Mol Neurobiol 2020; 40:813-828. [PMID: 31845161 PMCID: PMC11448950 DOI: 10.1007/s10571-019-00776-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/09/2019] [Indexed: 12/18/2022]
Abstract
In the present study, the functional role of the inwardly rectifying K+ channel, Kir4.1, and large-conductance Ca2+-activated K+ (BK) channel during cell migration in U251 cell line was investigated. We focused on polarised cells which are positive for the active-Cdc42 migration marker. The perforated patch technique was used to avoid intracellular dialysis and to maintain physiological changes in intracellular calcium. Wound healing was employed to assay migration after 24 h. Polarised cells recorded displayed different hallmarks of undifferentiated glial cells: depolarised resting membrane potential and high membrane resistance. Cells recorded outside wounded area did not display either constitutive inward or outward rectification. After migration, U251 cells were characterised by a constitutively smaller Kir4.1 and larger BK currents with a linearly related amplitude. Menthol modulation increased both currents in a linearly dependent manner, indicating a common mechanism triggered by activation of transient receptor potential melastatin 8 (TRPM8), a Ca2+-permeable non-selective cation channel. We hypothesised that both migration and menthol modulation would share an increase of intracellular calcium triggering the increase in Kir4.1 and BK channels. Immunocytochemistry demonstrated the cytoplasmic expression of both Kir4.1 and BK channels and a mislocation in the nucleus under basal conditions. Before and after migration, polarised cells increased the expression of Kir4.1 and BK channels both in the cytoplasm and nucleus. TEM ultrastructural analysis displayed a different nuclear distribution of Kir4.1 and BK channels. In the present study, the physiological role of Kir4.1 and BK currents at membrane potential, their involvement in migration, and the functional role of nuclear channels were discussed.
Collapse
Affiliation(s)
- Daniela Ratto
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Beatrice Ferrari
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
- Toxicology Unit, Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, ICS Maugeri SpA, IRCCS Pavia, 27100, Pavia, Italy
| | - Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, 1211, Geneva, Switzerland
| | - Stella Siciliani
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Carmine Di Iorio
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Filippo Cobelli
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Paola Veneroni
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy.
| |
Collapse
|
18
|
Liu J, Qu C, Han C, Chen MM, An LJ, Zou W. Potassium channels and their role in glioma: A mini review. Mol Membr Biol 2020; 35:76-85. [PMID: 32067536 DOI: 10.1080/09687688.2020.1729428] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
K+ channels regulate a multitude of biological processes and play important roles in a variety of diseases by controlling potassium flow across cell membranes. They are widely expressed in the central and peripheral nervous system. As a malignant tumor derived from nerve epithelium, glioma has the characteristics of high incidence, high recurrence rate, high mortality rate, and low cure rate. Since glioma cells show invasive growth, current surgical methods cannot completely remove tumors. Adjuvant chemotherapy is still needed after surgery. Because the blood-brain barrier and other factors lead to a lower effective concentration of chemotherapeutic drugs in the tumor, the recurrence rate of residual lesions is extremely high. Therefore, new therapeutic methods are needed. Numerous studies have shown that different K+ channel subtypes are differentially expressed in glioma cells and are involved in the regulation of the cell cycle of glioma cells to arrest them at different stages of the cell cycle. Increasing evidence suggests that K+ channels express in glioma cells and regulate glioma cell behaviors such as cell cycle, proliferation and apoptosis. This review article aims to summarize the current knowledge on the function of K+ channels in glioma, suggests K+ channels participating in the development of glioma.
Collapse
Affiliation(s)
- Jia Liu
- School of Life Science and Biotechnology, Faculty of Chemical, Environmental and Biological Science, Technology, Dalian University of Technology, Dalian, China.,College of Life Science, Liaoning Normal University, Dalian, China
| | - Chao Qu
- College of Life Science, Liaoning Normal University, Dalian, China
| | - Chao Han
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Meng-Meng Chen
- Company of Qingdao Re-Store Life Sciences, Qingdao, China
| | - Li-Jia An
- School of Life Science and Biotechnology, Faculty of Chemical, Environmental and Biological Science, Technology, Dalian University of Technology, Dalian, China
| | - Wei Zou
- College of Life Science, Liaoning Normal University, Dalian, China.,Company of Qingdao Re-Store Life Sciences, Qingdao, China
| |
Collapse
|
19
|
Zhang H, Liu Y, Men H, Zhang F, Zhang H. LRRCA8A and ANO1 contribute to serum-induced VRAC in a Ca 2+-dependent manners. J Pharmacol Sci 2020; 143:176-181. [PMID: 32386905 DOI: 10.1016/j.jphs.2020.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/05/2020] [Accepted: 03/23/2020] [Indexed: 10/24/2022] Open
Abstract
The volume-regulated anion channel (VRAC) plays a central role in maintaining cell volume in response to osmotic stress. Leucine-rich repeat-containing 8A (LRRC8A) was recently identified as an essential component of VRAC although other Cl- channels were also suggested to contribute to VRAC. VRAC is activated when a cell is challenged with a hypotonic environment or even in isotonic conditions challenged with different stimuli. It is not clear how VRAC is activated and whether activation of VRAC in hypotonic and isotonic conditions share the same mechanism. In this present study, we investigated relative contribution of LRRC8A and anoctamin 1(ANO1) to VRAC currents activated by fetal bovine serum (FBS) in isotonic condition, and studied the role of intracellular Ca2+ in this activation. We used CRISPR/Cas9 gene editing approach, electrophysiology, and pharmacology approaches to show that VRAC currents induced by FBS is mostly mediated by LRRC8A in HEK293 cells, but also with significant contribution from ANO1. FBS induces Ca2+ transients and these Ca2+ signals are required for the activation of VRAC by serum. These findings will help to further understand the mechanism in activation of VRAC.
Collapse
Affiliation(s)
- Huiran Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, Hebei, China; Department of Biopharmacy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yani Liu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China; Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong, China
| | - Hongchao Men
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China
| | - Fan Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
20
|
Yool AJ, Ramesh S. Molecular Targets for Combined Therapeutic Strategies to Limit Glioblastoma Cell Migration and Invasion. Front Pharmacol 2020; 11:358. [PMID: 32292341 PMCID: PMC7118801 DOI: 10.3389/fphar.2020.00358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
The highly invasive nature of glioblastoma imposes poor prospects for patient survival. Molecular evidence indicates glioblastoma cells undergo an intriguing expansion of phenotypic properties to include neuron-like signaling using excitable membrane ion channels and synaptic proteins, augmenting survival and motility. Neurotransmitter receptors, membrane signaling, excitatory receptors, and Ca2+ responses are important candidates for the design of customized treatments for cancers within the heterogeneous central nervous system. Relatively few published studies of glioblastoma multiforme (GBM) have evaluated pharmacological agents targeted to signaling pathways in limiting cancer cell motility. Transcriptomic analyses here identified classes of ion channels, ionotropic receptors, and synaptic proteins that are enriched in human glioblastoma biopsy samples. The pattern of GBM-enriched gene expression points to a major role for glutamate signaling. However, the predominant role of AMPA receptors in fast excitatory signaling throughout the central nervous system raises a challenge on how to target inhibitors selectively to cancer cells while maintaining tolerability. This review critically evaluates a panel of ligand- and voltage-gated ion channels and synaptic proteins upregulated in GBM, and the evidence for their potential roles in the pathological disease progress. Evidence suggests combinations of therapies could be more effective than single agents alone. Natural plant products used in traditional medicines for the treatment of glioblastoma contain flavonoids, terpenoids, polyphenols, epigallocatechin gallate, quinones, and saponins, which might serendipitously include agents that modulate some classes of signaling compounds highlighted in this review. New therapeutic strategies are likely to exploit evidence-based combinations of selected agents, each at a low dose, to create new cancer cell-specific therapeutics.
Collapse
Affiliation(s)
- Andrea J. Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sunita Ramesh
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
21
|
Echeverry S, Grismaldo A, Sánchez C, Sierra C, Henao JC, Granados ST, Sutachán JJ, Torres YP. Activation of BK Channel Contributes to PL-Induced Mesenchymal Stem Cell Migration. Front Physiol 2020; 11:210. [PMID: 32265729 PMCID: PMC7105713 DOI: 10.3389/fphys.2020.00210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/24/2020] [Indexed: 01/16/2023] Open
Abstract
Due to their capacity to proliferate, migrate, and differentiate, mesenchymal stem cells (MSCs) are considered to be good candidates for regenerative medicine applications. The mechanisms underlying proliferation and differentiation of MSCs have been studied. However, much less is known about the mechanisms regulating the migration of MSCs. Platelet lysate (PL), a supplement used to promote cell expansion, has been shown to promote MSCs migration; however, the underlying mechanism are unknown. Here, by using adipose-derived rat MSCs (rMSCs) and the scratch assay in the absence and presence of various BK channels modulators, we evaluated the role of BK channels in mediating the PL-stimulated migration of rMSCs. We found that 5% PL increased rMSCs migration, and this effect was blocked by the addition of the BK channel selective antagonist Iberiotoxin (IBTX). In the absence of PL, the BK channel agonist NS1619, stimulated rMSCs migration to similar level as 5% PL. Addition of both NS1619 and 5% PL resulted in an increase in rMSCs migration, that was higher than when either one was added individually. From whole-cell recordings, it was found that the addition of 5% PL increased the magnitude of BK current density. By using Western blot and flow cytometry, it was found that PL did not affect the expression of BK channels. Together, our results indicate that as shown in other cell types, activation of BK channels by themselves also promote rMSC migration, and show that activation of BK channels contribute to the observed PL-induced increase in migration of rMSC.
Collapse
|
22
|
Ragonese F, Monarca L, Bastioli F, Arcuri C, Mancinelli L, Fioretti B. Silver ions promote blebs growth in U251 glioblastoma cell by activating nonselective cationic currents. Sci Rep 2019; 9:12898. [PMID: 31501459 PMCID: PMC6733836 DOI: 10.1038/s41598-019-49198-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/21/2019] [Indexed: 11/09/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive human brain cancer with low prognosis and therefore the discovery of new anticancer agents is needful. Sulfydryl reagents, such as silver, have been shown to induce membrane vesiculation in several cellular models through a mechanism that has not been yet completely clarified. Using U251 glioblastoma cells, we observed that silver induced irreversible bleb formation of the plasma membrane. This morphological event was anticipated by an increase of intracellular Ca2+ associated to extracellular Ca2+ influx. Accordingly, using patch-clamp whole cell recording during silver ion application, inward current/s (IAg) at -90 mV were detected and cells were permeable to Ca2+ and monovalent ions such as Na+. IAg activation and the intracellular Ca2+ increase promoted by silver ions (Ag+) were prevented by co-application of 20 µM cysteine and 300 µM DIDS (4,4'-Diisothiocyanatostilbene-2,2'-disulfonic acid), suggesting a critical role of thiol groups in the biological effects of silver ions. IAg was partially inhibited by 1 mM Gd3+, an unspecific inhibitor of cationic currents. Cysteine, Gd3+ and extracellular free Ca2+ solution completely abolished blebbing formation promoted by Ag+. Furthermore, extracellular Na+ ion replacement with TEA or an increase of extracellular tonicity by sucrose (100 mM) reduced both size and growth of membrane blebbing. Our data suggest that Ag+ promotes the formation necrotic blebs as consequence of the increase of intracellular Ca2+ and intracellular hydrostatic pressure associated to the activation of cationic currents. Since silver-induced blebs were less evident in benign glial human Müller MIO-M1 cells, silver compounds could represent new adjuvant to anticancer agents to improve GBM therapies.
Collapse
Affiliation(s)
- Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, Via Elce di Sotto 8, University of Perugia, Perugia, Italy.,Department of Experimental Medicine, Piazzale Gambuli 1, University of Perugia, Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, Via Elce di Sotto 8, University of Perugia, Perugia, Italy
| | - Federica Bastioli
- Department of Chemistry, Biology and Biotechnologies, Via Elce di Sotto 8, University of Perugia, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Piazzale Gambuli 1, University of Perugia, Perugia, Italy
| | - Loretta Mancinelli
- Department of Chemistry, Biology and Biotechnologies, Via Elce di Sotto 8, University of Perugia, Perugia, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, Via Elce di Sotto 8, University of Perugia, Perugia, Italy.
| |
Collapse
|
23
|
Saberbaghi T, Wong R, Rutka JT, Wang GL, Feng ZP, Sun HS. Role of Cl− channels in primary brain tumour. Cell Calcium 2019; 81:1-11. [DOI: 10.1016/j.ceca.2019.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/28/2019] [Accepted: 05/13/2019] [Indexed: 12/20/2022]
|
24
|
Caramia M, Sforna L, Franciolini F, Catacuzzeno L. The Volume-Regulated Anion Channel in Glioblastoma. Cancers (Basel) 2019; 11:cancers11030307. [PMID: 30841564 PMCID: PMC6468384 DOI: 10.3390/cancers11030307] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 01/02/2023] Open
Abstract
Malignancy of glioblastoma multiforme (GBM), the most common and aggressive form of human brain tumor, strongly depends on its enhanced cell invasion and death evasion which make surgery and accompanying therapies highly ineffective. Several ion channels that regulate membrane potential, cytosolic Ca2+ concentration and cell volume in GBM cells play significant roles in sustaining these processes. Among them, the volume-regulated anion channel (VRAC), which mediates the swelling-activated chloride current (IClswell) and is highly expressed in GBM cells, arguably plays a major role. VRAC is primarily involved in reestablishing the original cell volume that may be lost under several physiopathological conditions, but also in sustaining the shape and cell volume changes needed for cell migration and proliferation. While experimentally VRAC is activated by exposing cells to hypotonic solutions that cause the increase of cell volume, in vivo it is thought to be controlled by several different stimuli and modulators. In this review we focus on our recent work showing that two conditions normally occurring in pathological GBM tissues, namely high serum levels and severe hypoxia, were both able to activate VRAC, and their activation was found to promote cell migration and resistance to cell death, both features enhancing GBM malignancy. Also, the fact that the signal transduction pathway leading to VRAC activation appears to involve GBM specific intracellular components, such as diacylglicerol kinase and phosphatidic acid, reportedly not involved in the activation of VRAC in healthy tissues, is a relevant finding. Based on these observations and the impact of VRAC in the physiopathology of GBM, targeting this channel or its intracellular regulators may represent an effective strategy to contrast this lethal tumor.
Collapse
Affiliation(s)
- Martino Caramia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy.
| | - Luigi Sforna
- Department of Experimental Medicine, University of Perugia, Perugia 06132, Italy.
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy.
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy.
| |
Collapse
|
25
|
Cancer-Associated Intermediate Conductance Ca 2+-Activated K⁺ Channel K Ca3.1. Cancers (Basel) 2019; 11:cancers11010109. [PMID: 30658505 PMCID: PMC6357066 DOI: 10.3390/cancers11010109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Several tumor entities have been reported to overexpress KCa3.1 potassium channels due to epigenetic, transcriptional, or post-translational modifications. By modulating membrane potential, cell volume, or Ca2+ signaling, KCa3.1 has been proposed to exert pivotal oncogenic functions in tumorigenesis, malignant progression, metastasis, and therapy resistance. Moreover, KCa3.1 is expressed by tumor-promoting stroma cells such as fibroblasts and the tumor vasculature suggesting a role of KCa3.1 in the adaptation of the tumor microenvironment. Combined, this features KCa3.1 as a candidate target for innovative anti-cancer therapy. However, immune cells also express KCa3.1 thereby contributing to T cell activation. Thus, any strategy targeting KCa3.1 in anti-cancer therapy may also modulate anti-tumor immune activity and/or immunosuppression. The present review article highlights the potential of KCa3.1 as an anti-tumor target providing an overview of the current knowledge on its function in tumor pathogenesis with emphasis on vasculo- and angiogenesis as well as anti-cancer immune responses.
Collapse
|
26
|
Role of KCa3.1 Channels in Modulating Ca 2+ Oscillations during Glioblastoma Cell Migration and Invasion. Int J Mol Sci 2018; 19:ijms19102970. [PMID: 30274242 PMCID: PMC6213908 DOI: 10.3390/ijms19102970] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 01/29/2023] Open
Abstract
Cell migration and invasion in glioblastoma (GBM), the most lethal form of primary brain tumors, are critically dependent on Ca2+ signaling. Increases of [Ca2+]i in GBM cells often result from Ca2+ release from the endoplasmic reticulum (ER), promoted by a variety of agents present in the tumor microenvironment and able to activate the phospholipase C/inositol 1,4,5-trisphosphate PLC/IP3 pathway. The Ca2+ signaling is further strengthened by the Ca2+ influx from the extracellular space through Ca2+ release-activated Ca2+ (CRAC) currents sustained by Orai/STIM channels, meant to replenish the partially depleted ER. Notably, the elevated cytosolic [Ca2+]i activates the intermediate conductance Ca2+-activated K (KCa3.1) channels highly expressed in the plasma membrane of GBM cells, and the resulting K+ efflux hyperpolarizes the cell membrane. This translates to an enhancement of Ca2+ entry through Orai/STIM channels as a result of the increased electromotive (driving) force on Ca2+ influx, ending with the establishment of a recurrent cycle reinforcing the Ca2+ signal. Ca2+ signaling in migrating GBM cells often emerges in the form of intracellular Ca2+ oscillations, instrumental to promote key processes in the migratory cycle. This has suggested that KCa3.1 channels may promote GBM cell migration by inducing or modulating the shape of Ca2+ oscillations. In accordance, we recently built a theoretical model of Ca2+ oscillations incorporating the KCa3.1 channel-dependent dynamics of the membrane potential, and found that the KCa3.1 channel activity could significantly affect the IP3 driven Ca2+ oscillations. Here we review our new theoretical model of Ca2+ oscillations in GBM, upgraded in the light of better knowledge of the KCa3.1 channel kinetics and Ca2+ sensitivity, the dynamics of the Orai/STIM channel modulation, the migration and invasion mechanisms of GBM cells, and their regulation by Ca2+ signals.
Collapse
|
27
|
D'Alessandro G, Limatola C, Catalano M. Functional Roles of the Ca2+-activated K+ Channel, KCa3.1, in Brain Tumors. Curr Neuropharmacol 2018; 16:636-643. [PMID: 28707595 PMCID: PMC5997864 DOI: 10.2174/0929867324666170713103621] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/22/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Glioblastoma is the most aggressive and deadly brain tumor, with low disease-free period even after surgery and combined radio and chemotherapies. Among the factors contributing to the devastating effect of this tumor in the brain are the elevated proliferation and invasion rate, and the ability to induce a local immunosuppressive environment. The intermediateconductance Ca2+-activated K+ channel KCa3.1 is expressed in glioblastoma cells and in tumorinfiltrating cells. METHODS We first describe the researches related to the role of KCa3.1 channels in the invasion of brain tumor cells and the regulation of cell cycle. In the second part we review the involvement of KCa3.1 channel in tumor-associated microglia cell behaviour. RESULTS In tumor cells, the functional expression of KCa3.1 channels is important to substain cell invasion and proliferation. In tumor infiltrating cells, KCa3.1 channel activity is required to regulate their activation state. Interfering with KCa3.1 activity can be an adjuvant therapeutic approach in addition to classic chemotherapy and radiotherapy, to counteract tumor growth and prolong patient's survival. CONCLUSION In this mini-review we discuss the evidence of the functional roles of KCa3.1 channels in glioblastoma biology.
Collapse
Affiliation(s)
- Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
28
|
Rosa P, Catacuzzeno L, Sforna L, Mangino G, Carlomagno S, Mincione G, Petrozza V, Ragona G, Franciolini F, Calogero A. BK channels blockage inhibits hypoxia-induced migration and chemoresistance to cisplatin in human glioblastoma cells. J Cell Physiol 2018; 233:6866-6877. [PMID: 29319175 DOI: 10.1002/jcp.26448] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) cells express large-conductance, calcium-activated potassium (BK) channels, whose activity is important for several critical aspects of the tumor, such as migration/invasion and cell death. GBMs are also characterized by a heavy hypoxic microenvironment that exacerbates tumor aggressiveness. Since hypoxia modulates the activity of BK channels in many tissues, we hypothesized that a hypoxia-induced modulation of these channels may contribute to the hypoxia-induced GBM aggressiveness. In U87-MG cells, hypoxia induced a functional upregulation of BK channel activity, without interfering with their plasma membrane expression. Wound healing and transwell migration assays showed that hypoxia increased the migratory ability of U87-MG cells, an effect that could be prevented by BK channel inhibition. Toxicological experiments showed that hypoxia was able to induce chemoresistance to cisplatin in U87-MG cells and that the inhibition of BK channels prevented the hypoxia-induced chemoresistance. Clonogenic assays showed that BK channels are also used to increase the clonogenic ability of U87-MG GBM cells in presence, but not in absence, of cisplatin. BK channels were also found to be essential for the hypoxia-induced de-differentiation of GBM cells. Finally, using immunohistochemical analysis, we highlighted the presence of BK channels in hypoxic areas of human GBM tissues, suggesting that our findings may have physiopathological relevance in vivo. In conclusion, our data show that BK channels promote several aspects of the aggressive potential of GBM cells induced by hypoxia, such as migration and chemoresistance to cisplatin, suggesting it as a potential therapeutic target in the treatment of GBM.
Collapse
Affiliation(s)
- Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Giorgio Mangino
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Silvia Carlomagno
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Gabriella Mincione
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara, Italy
| | - Vincenzo Petrozza
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| | - Giuseppe Ragona
- Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy.,Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| |
Collapse
|
29
|
Reactive Astrocytes in Glioblastoma Multiforme. Mol Neurobiol 2018; 55:6927-6938. [PMID: 29363044 DOI: 10.1007/s12035-018-0880-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/07/2018] [Indexed: 12/17/2022]
Abstract
Despite the multidisciplinary integration in the therapeutic management of glioblastoma multiforme (GBM), the prognosis of GBM patients is poor. There is growing recognition that the cells in the tumor microenvironment play a vital role in regulating the progression of glioma. Astrocytes are an important component of the blood-brain barrier (BBB) as well as the tripartite synapse neural network to promote bidirectional communication with neurons under physiological conditions. Emerging evidence shows that tumor-associated reactive astrocytes interact with glioma cells and facilitate the progression, aggression, and survival of tumors by releasing different cytokines. Communication between reactive astrocytes and glioma cells is further promoted through ion channels and ion transporters, which augment the migratory capacity and invasiveness of tumor cells by modifying H+ and Ca2+ concentrations and stimulating volume changes in the cell. This in part contributes to the loss of epithelial polarization, initiating epithelial-mesenchymal transition. Therefore, this review will summarize the recent findings on the role of reactive astrocytes in the progression of GBM and in the development of treatment-resistant glioma. In addition, the involvement of ion channels and transporters in bridging the interactions between tumor cells and astrocytes and their potential as new therapeutic anti-tumor targets will be discussed.
Collapse
|
30
|
Klumpp L, Sezgin EC, Skardelly M, Eckert F, Huber SM. KCa3.1 Channels and Glioblastoma: In Vitro Studies. Curr Neuropharmacol 2018; 16:627-635. [PMID: 28786347 PMCID: PMC5997865 DOI: 10.2174/1570159x15666170808115821] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several tumor entities including brain tumors aberrantly overexpress intermediate conductance Ca2+ activated KCa3.1 K+ channels. These channels contribute significantly to the transformed phenotype of the tumor cells. METHOD PubMed was searched in order to summarize our current knowledge on the molecular signaling upstream and downstream and the effector functions of KCa3.1 channel activity in tumor cells in general and in glioblastoma cells in particular. In addition, KCa3.1 expression and function for repair of DNA double strand breaks was determined experimentally in primary glioblastoma cultures in dependence on the abundance of proneural and mesenchymal stem cell markers. RESULTS By modulating membrane potential, cell volume, Ca2+ signals and the respiratory chain, KCa3.1 channels in both, plasma and inner mitochondrial membrane, have been demonstrated to regulate many cellular processes such as migration and tissue invasion, metastasis, cell cycle progression, oxygen consumption and metabolism, DNA damage response and cell death of cancer cells. Moreover, KCa3.1 channels have been shown to crucially contribute to resistance against radiotherapy. Futhermore, the original in vitro data on KCa3.1 channel expression in subtypes of glioblastoma stem(-like) cells propose KCa3.1 as marker for the mesenchymal subgroup of cancer stem cells and suggest that KCa3.1 contributes to the therapy resistance of mesenchymal glioblastoma stem cells. CONCLUSION The data suggest KCa3.1 channel targeting in combination with radiotherapy as promising new tool to eradicate therapy-resistant mesenchymal glioblastoma stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Stephan M. Huber
- Address correspondence to this author at the Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; Tel: +49-(0)7071-29-82183; E-mail:
| |
Collapse
|
31
|
Sforna L, Megaro A, Pessia M, Franciolini F, Catacuzzeno L. Structure, Gating and Basic Functions of the Ca2+-activated K Channel of Intermediate Conductance. Curr Neuropharmacol 2018; 16:608-617. [PMID: 28875832 PMCID: PMC5997868 DOI: 10.2174/1570159x15666170830122402] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/21/2017] [Accepted: 07/22/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The KCa3.1 channel is the intermediate-conductance member of the Ca2+- activated K channel superfamily. It is widely expressed in excitable and non-excitable cells, where it plays a major role in a number of cell functions. This paper aims at illustrating the main structural, biophysical and modulatory properties of the KCa3.1 channel, and providing an account of experimental data on its role in volume regulation and Ca2+ signals. METHODS Research and online content related to the structure, structure/function relationship, and physiological role of the KCa3.1 channel are reviewed. RESULTS Expressed in excitable and non-excitable cells, the KCa3.1 channel is voltage independent, its opening being exclusively gated by the binding of intracellular Ca2+ to calmodulin, a Ca2+- binding protein constitutively associated with the C-terminus of each KCa3.1 channel α subunit. The KCa3.1 channel activates upon high affinity Ca2+ binding, and in highly coordinated fashion giving steep Hill functions and relatively low EC50 values (100-350 nM). This high Ca2+ sensitivity is physiologically modulated by closely associated kinases and phosphatases. The KCa3.1 channel is normally activated by global Ca2+ signals as resulting from Ca2+ released from intracellular stores, or by the refilling influx through store operated Ca2+ channels, but cases of strict functional coupling with Ca2+-selective channels are also found. KCa3.1 channels are highly expressed in many types of cells, where they play major roles in cell migration and death. The control of these complex cellular processes is achieved by KCa3.1 channel regulation of the driving force for Ca2+ entry from the extracellular medium, and by mediating the K+ efflux required for cell volume control. CONCLUSION Much work remains to be done to fully understand the structure/function relationship of the KCa3.1 channels. Hopefully, this effort will provide the basis for a beneficial modulation of channel activity under pathological conditions.
Collapse
Affiliation(s)
| | | | | | - Fabio Franciolini
- Address correspondence to these authors at the Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Pascoli, 8-06123, Perugia; Tel: 39.075.585.5751; E-mails: and
| | - Luigi Catacuzzeno
- Address correspondence to these authors at the Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Pascoli, 8-06123, Perugia; Tel: 39.075.585.5751; E-mails: and
| |
Collapse
|
32
|
Lu FF, Wang HY, He XZ, Liang TY, Wang W, Hu HM, Wu F, Liu YW, Zhang SZ. Prognostic value of ion channel genes in Chinese patients with gliomas based on mRNA expression profiling. J Neurooncol 2017; 134:397-405. [DOI: 10.1007/s11060-017-2539-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 06/25/2017] [Indexed: 01/17/2023]
|
33
|
Rosa P, Sforna L, Carlomagno S, Mangino G, Miscusi M, Pessia M, Franciolini F, Calogero A, Catacuzzeno L. Overexpression of Large-Conductance Calcium-Activated Potassium Channels in Human Glioblastoma Stem-Like Cells and Their Role in Cell Migration. J Cell Physiol 2017; 232:2478-2488. [PMID: 27606467 DOI: 10.1002/jcp.25592] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/07/2016] [Indexed: 01/24/2023]
Abstract
Glioblastomas (GBMs) are brain tumors characterized by diffuse invasion of cancer cells into the healthy brain parenchyma, and establishment of secondary foci. GBM cells abundantly express large-conductance, calcium-activated potassium (BK) channels that are thought to promote cell invasion. Recent evidence suggests that the GBM high invasive potential mainly originates from a pool of stem-like cells, but the expression and function of BK channels in this cell subpopulation have not been studied. We investigated the expression of BK channels in GBM stem-like cells using electrophysiological and immunochemical techniques, and assessed their involvement in the migratory process of this important cell subpopulation. In U87-MG cells, BK channel expression and function were markedly upregulated by growth conditions that enriched the culture in GBM stem-like cells (U87-NS). Cytofluorimetric analysis further confirmed the appearance of a cell subpopulation that co-expressed high levels of BK channels and CD133, as well as other stem cell markers. A similar association was also found in cells derived from freshly resected GBM biopsies. Finally, transwell migration tests showed that U87-NS cells migration was much more sensitive to BK channel block than U87-MG cells. Our data show that BK channels are highly expressed in GBM stem-like cells, and participate to their high migratory activity. J. Cell. Physiol. 232: 2478-2488, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paolo Rosa
- Department of Medico-Surgical Sciences and Biotechnologies, "La Sapienza" University, Latina, Italy
| | - Luigi Sforna
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Silvia Carlomagno
- Department of Medico-Surgical Sciences and Biotechnologies, "La Sapienza" University, Latina, Italy
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, "La Sapienza" University, Latina, Italy
| | - Massimo Miscusi
- Department of Medico-Surgical Sciences and Biotechnologies, "La Sapienza" University, Latina, Italy
| | - Mauro Pessia
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Fabio Franciolini
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Antonella Calogero
- Department of Medico-Surgical Sciences and Biotechnologies, "La Sapienza" University, Latina, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
34
|
KCa3.1 (IK) modulates pancreatic cancer cell migration, invasion and proliferation: anomalous effects on TRAM-34. Pflugers Arch 2016; 468:1865-1875. [DOI: 10.1007/s00424-016-1891-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 12/30/2022]
|
35
|
Sforna L, Cenciarini M, Belia S, Michelucci A, Pessia M, Franciolini F, Catacuzzeno L. Hypoxia Modulates the Swelling-Activated Cl Current in Human Glioblastoma Cells: Role in Volume Regulation and Cell Survival. J Cell Physiol 2016; 232:91-100. [PMID: 27028592 DOI: 10.1002/jcp.25393] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/25/2016] [Indexed: 12/18/2022]
Abstract
The malignancy of glioblastoma multiforme (GBM), the most common human brain tumor, correlates with the presence of hypoxic areas, but the underlying mechanisms are unclear. GBM cells express abundant Cl channels whose activity supports cell volume and membrane potential changes, ultimately leading to cell proliferation, migration, and escaping death. In non-tumor tissues Cl channels are modulated by hypoxia, which prompted us to verify whether hypoxia would also modulate Cl channels in GBM cells. Our results show that in GBM cell lines, acute application of a hypoxic solution activates a Cl current displaying the biophysical and pharmacological features of the swelling-activated Cl current (ICl,swell ). We also found that acute hypoxia increased the cell volume by about 20%, and a 30% hypertonic solution partially inhibited the hypoxia-activated Cl current, suggesting that cell swelling and the activation of the Cl current are sequential events. Notably, the hypoxia-induced cell swelling was followed by a regulatory volume decrease (RVD) mediated mainly by ICl,swell . Since, a hypoxia-induced prolonged cell swelling is usually regarded as a death insult, we hypothesized that the hypoxia-activated Cl current could limit cell swelling and prevent necrotic death of GBM cells under hypoxic conditions. In accordance, we found that the ICl,swell inhibitor DCPIB hampered the RVD process, and more importantly it sensibly increased the hypoxia-induced necrotic death in these cells. Taken together, these results suggest that Cl channels are strongly involved in the survival of GBM cells in a hypoxic environment, and may thus represent a new therapeutic target for this malignant tumor. J. Cell. Physiol. 232: 91-100, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.,Department of Experimental Medicine, University of Perugia, Italy
| | - Marta Cenciarini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy
| | - Antonio Michelucci
- Department of Neuroscience, Imaging and Clinical Sciences, University of Chieti 'G. d'Annunzio', Italy
| | - Mauro Pessia
- Department of Experimental Medicine, University of Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.
| |
Collapse
|
36
|
Stegen B, Klumpp L, Misovic M, Edalat L, Eckert M, Klumpp D, Ruth P, Huber SM. K + channel signaling in irradiated tumor cells. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:585-598. [PMID: 27165704 DOI: 10.1007/s00249-016-1136-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/24/2016] [Accepted: 04/20/2016] [Indexed: 12/17/2022]
Abstract
K+ channels crosstalk with biochemical signaling cascades and regulate virtually all cellular processes by adjusting the intracellular K+ concentration, generating the membrane potential, mediating cell volume changes, contributing to Ca2+ signaling, and directly interacting within molecular complexes with membrane receptors and downstream effectors. Tumor cells exhibit aberrant expression and activity patterns of K+ channels. The upregulation of highly "oncogenic" K+ channels such as the Ca2+-activated IK channel may drive the neoplastic transformation, malignant progression, metastasis, or therapy resistance of tumor cells. In particular, ionizing radiation in doses used for fractionated radiotherapy in the clinic has been shown to activate K+ channels. Radiogenic K+ channel activity, in turn, contributes to the DNA damage response and promotes survival of the irradiated tumor cells. Tumor-specific overexpression of certain K+ channel types together with the fact that pharmacological K+ channel modulators are already in clinical use or well tolerated in clinical trials suggests that K+ channel targeting alone or in combination with radiotherapy might become a promising new strategy of anti-cancer therapy. The present article aims to review our current knowledge on K+ channel signaling in irradiated tumor cells. Moreover, it provides new data on molecular mechanisms of radiogenic K+ channel activation and downstream signaling events.
Collapse
Affiliation(s)
- Benjamin Stegen
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Milan Misovic
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Lena Edalat
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Marita Eckert
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Dominik Klumpp
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
37
|
Huber SM, Butz L, Stegen B, Klumpp L, Klumpp D, Eckert F. Role of ion channels in ionizing radiation-induced cell death. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2657-64. [DOI: 10.1016/j.bbamem.2014.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/30/2014] [Accepted: 11/05/2014] [Indexed: 02/05/2023]
|
38
|
Stegen B, Butz L, Klumpp L, Zips D, Dittmann K, Ruth P, Huber SM. Ca2+-Activated IK K+ Channel Blockade Radiosensitizes Glioblastoma Cells. Mol Cancer Res 2015; 13:1283-95. [PMID: 26041939 DOI: 10.1158/1541-7786.mcr-15-0075] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/22/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Ca(2+)-activated K(+) channels, such as BK and IK channels, have been proposed to fulfill pivotal functions in neoplastic transformation, malignant progression, and brain infiltration of glioblastoma cells. Here, the ionizing radiation (IR) effect of IK K(+) channel targeting was tested in human glioblastoma cells. IK channels were inhibited pharmacologically by TRAM-34 or genetically by knockdown, cells were irradiated with 6 MV photons and IK channel activity, Ca(2+) signaling, cell cycling, residual double-strand breaks, and clonogenic survival were determined. In addition, the radiosensitizing effect of TRAM-34 was analyzed in vivo in ectopic tumors. Moreover, The Cancer Genome Atlas (TCGA) was queried to expose the dependence of IK mRNA abundance on overall survival (OS) of patients with glioma. Results indicate that radiation increased the activity of IK channels, modified Ca(2+) signaling, and induced a G2-M cell-cycle arrest. TRAM-34 decreased the IR-induced accumulation in G2-M arrest and increased the number of γH2AX foci post-IR, suggesting that TRAM-34 mediated an increase of residual DNA double-strand breaks. Mechanistically, IK knockdown abolished the TRAM-34 effects indicating the IK specificity of TRAM-34. Finally, TRAM-34 radiosensitized ectopic glioblastoma in vivo and high IK mRNA abundance associated with shorter patient OS in low-grade glioma and glioblastoma. IMPLICATIONS Together, these data support a cell-cycle regulatory function for IK K(+) channels, and combined therapy using IK channel targeting and radiation is a new strategy for anti-glioblastoma therapy.
Collapse
Affiliation(s)
- Benjamin Stegen
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Lena Butz
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany. Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany. Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Dittmann
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
39
|
Catacuzzeno L, Caramia M, Sforna L, Belia S, Guglielmi L, D'Adamo MC, Pessia M, Franciolini F. Reconciling the discrepancies on the involvement of large-conductance Ca(2+)-activated K channels in glioblastoma cell migration. Front Cell Neurosci 2015; 9:152. [PMID: 25941475 PMCID: PMC4403502 DOI: 10.3389/fncel.2015.00152] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/02/2015] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor, and is notable for spreading so effectively through the brain parenchyma to make complete surgical resection virtually impossible, and prospect of life dismal. Several ion channels have been involved in GBM migration and invasion, due to their critical role in supporting volume changes and Ca(2+) influx occuring during the process. The large-conductance, Ca(2+)-activated K (BK) channels, markedly overexpressed in biopsies of patients with GBMs and in GBM cell lines, have attracted much interest and have been suggested to play a central role in cell migration and invasion as candidate channels for providing the ion efflux and consequent water extrusion that allow cell shrinkage during migration. Available experimental data on the role of BK channel in migration and invasion are not consistent though. While BK channels block typically resulted in inhibition of cell migration or in no effect, their activation would either enhance or inhibit the process. This short review reexamines the relevant available data on the topic, and presents a unifying paradigm capable of reconciling present discrepancies. According to this paradigm, BK channels would not contribute to migration under conditions where the [Ca(2+)] i is too low for their activation. They will instead positively contribute to migration for intermediate [Ca(2+)] i , insufficient as such to activate BK channels, but capable of predisposing them to cyclic activation following oscillatory [Ca(2+)] i increases. Finally, steadily active BK channels because of prolonged high [Ca(2+)] i would inhibit migration as their steady activity would be unsuitable to match the cyclic cell volume changes needed for proper cell migration.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Dipartimento di Chimica, Biologia e Biotecnologie, Universita' di Perugia Perugia, Italy
| | - Martino Caramia
- Dipartimento di Chimica, Biologia e Biotecnologie, Universita' di Perugia Perugia, Italy
| | - Luigi Sforna
- Dipartimento di Chimica, Biologia e Biotecnologie, Universita' di Perugia Perugia, Italy
| | - Silvia Belia
- Dipartimento di Chimica, Biologia e Biotecnologie, Universita' di Perugia Perugia, Italy
| | - Luca Guglielmi
- Dipartimento di Medicina Sperimentale, Scuola di Medicina e Chirurgia, Universita' di Perugia Perugia, Italy
| | - Maria Cristina D'Adamo
- Dipartimento di Medicina Sperimentale, Scuola di Medicina e Chirurgia, Universita' di Perugia Perugia, Italy
| | - Mauro Pessia
- Dipartimento di Medicina Sperimentale, Scuola di Medicina e Chirurgia, Universita' di Perugia Perugia, Italy
| | - Fabio Franciolini
- Dipartimento di Chimica, Biologia e Biotecnologie, Universita' di Perugia Perugia, Italy
| |
Collapse
|
40
|
Litan A, Langhans SA. Cancer as a channelopathy: ion channels and pumps in tumor development and progression. Front Cell Neurosci 2015; 9:86. [PMID: 25852478 PMCID: PMC4362317 DOI: 10.3389/fncel.2015.00086] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/23/2015] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests that ion channels and pumps not only regulate membrane potential, ion homeostasis, and electric signaling in excitable cells but also play important roles in cell proliferation, migration, apoptosis and differentiation. Consistent with a role in cell signaling, channel proteins and ion pumps can form macromolecular complexes with growth factors, and cell adhesion and other signaling molecules. And while cancer is still not being cataloged as a channelopathy, as the non-traditional roles of ion pumps and channels are being recognized, it is increasingly being suggested that ion channels and ion pumps contribute to cancer progression. Cancer cell migration requires the regulation of adhesion complexes between migrating cells and surrounding extracellular matrix (ECM) proteins. Cell movement along solid surfaces requires a sequence of cell protrusions and retractions that mainly depend on regulation of the actin cytoskeleton along with contribution of microtubules and molecular motor proteins such as mysoin. This process is triggered and modulated by a combination of environmental signals, which are sensed and integrated by membrane receptors, including integrins and cadherins. Membrane receptors transduce these signals into downstream signaling pathways, often involving the Rho GTPase protein family. These pathways regulate the cytoskeletal rearrangements necessary for proper timing of adhesion, contraction and detachment of cells in order to find their way through extracellular spaces. Migration and adhesion involve continuous modulation of cell motility, shape and volume, in which ion channels and pumps play major roles. Research on cancer cells suggests that certain ion channels may be involved in aberrant tumor growth and channel inhibitors often lead to growth arrest. This review will describe recent research into the role of ion pumps and ion channels in cell migration and adhesion, and how they may contribute to tumor development.
Collapse
Affiliation(s)
- Alisa Litan
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children Wilmington, DE, USA
| | - Sigrid A Langhans
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children Wilmington, DE, USA
| |
Collapse
|
41
|
Sforna L, Cenciarini M, Belia S, D'Adamo MC, Pessia M, Franciolini F, Catacuzzeno L. The role of ion channels in the hypoxia-induced aggressiveness of glioblastoma. Front Cell Neurosci 2015; 8:467. [PMID: 25642170 PMCID: PMC4295544 DOI: 10.3389/fncel.2014.00467] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The malignancy of glioblastoma multiform (GBM), the most common and aggressive form of human brain tumors, strongly correlates with the presence of hypoxic areas, but the mechanisms controlling the hypoxia-induced aggressiveness are still unclear. GBM cells express a number of ion channels whose activity supports cell volume changes and increases in the cytosolic Ca2+ concentration, ultimately leading to cell proliferation, migration or death. In several cell types it has previously been shown that low oxygen levels regulate the expression and activity of these channels, and more recent data indicate that this also occurs in GBM cells. Based on these findings, it may be hypothesized that the modulation of ion channel activity or expression by the hypoxic environment may participate in the acquisition of the aggressive phenotype observed in GBM cells residing in a hypoxic environment. If this hypothesis will be confirmed, the use of available ion channels modulators may be considered for implementing novel therapeutic strategies against these tumors.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Marta Cenciarini
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Maria Cristina D'Adamo
- Faculty of Medicine, Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Mauro Pessia
- Faculty of Medicine, Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| |
Collapse
|
42
|
Turner KL, Honasoge A, Robert SM, McFerrin MM, Sontheimer H. A proinvasive role for the Ca(2+) -activated K(+) channel KCa3.1 in malignant glioma. Glia 2014; 62:971-81. [PMID: 24585442 DOI: 10.1002/glia.22655] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/03/2014] [Accepted: 02/14/2014] [Indexed: 11/09/2022]
Abstract
Glioblastoma multiforme are highly motile primary brain tumors. Diffuse tissue invasion hampers surgical resection leading to poor patient prognosis. Recent studies suggest that intracellular Ca(2+) acts as a master regulator for cell motility and engages a number of downstream signals including Ca(2+) -activated ion channels. Querying the REepository of Molecular BRAin Neoplasia DaTa (REMBRANDT), an annotated patient gene database maintained by the National Cancer Institute, we identified the intermediate conductance Ca(2+) -activated K(+) channels, KCa3.1, being overexpressed in 32% of glioma patients where protein expression significantly correlated with poor patient survival. To mechanistically link KCa3.1 expression to glioma invasion, we selected patient gliomas that, when propagated as xenolines in vivo, present with either high or low KCa3.1 expression. In addition, we generated U251 glioma cells that stably express an inducible knockdown shRNA to experimentally eliminate KCa3.1 expression. Subjecting these cells to a combination of in vitro and in situ invasion assays, we demonstrate that KCa3.1 expression significantly enhances glioma invasion and that either specific pharmacological inhibition with TRAM-34 or elimination of the channel impairs invasion. Importantly, after intracranial implantation into SCID mice, ablation of KCa3.1 with inducible shRNA resulted in a significant reduction in tumor invasion into surrounding brain in vivo. These results show that KCa3.1 confers an invasive phenotype that significantly worsens a patient's outlook, and suggests that KCa3.1 represents a viable therapeutic target to reduce glioma invasion.
Collapse
Affiliation(s)
- Kathryn L Turner
- Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, Alabama
| | | | | | | | | |
Collapse
|
43
|
Urrego D, Tomczak AP, Zahed F, Stühmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130094. [PMID: 24493742 PMCID: PMC3917348 DOI: 10.1098/rstb.2013.0094] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Normal cell-cycle progression is a crucial task for every multicellular organism, as it determines body size and shape, tissue renewal and senescence, and is also crucial for reproduction. On the other hand, dysregulation of the cell-cycle progression leading to uncontrolled cell proliferation is the hallmark of cancer. Therefore, it is not surprising that it is a tightly regulated process, with multifaceted and very complex control mechanisms. It is now well established that one of those mechanisms relies on ion channels, and in many cases specifically on potassium channels. Here, we summarize the possible mechanisms underlying the importance of potassium channels in cell-cycle control and briefly review some of the identified channels that illustrate the multiple ways in which this group of proteins can influence cell proliferation and modulate cell-cycle progression.
Collapse
Affiliation(s)
- Diana Urrego
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, , Hermann-Rein-Strasse 3, Göttingen 37075, Germany
| | | | | | | | | |
Collapse
|
44
|
Lang F, Stournaras C. Ion channels in cancer: future perspectives and clinical potential. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130108. [PMID: 24493756 DOI: 10.1098/rstb.2013.0108] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ion transport across the cell membrane mediated by channels and carriers participate in the regulation of tumour cell survival, death and motility. Moreover, the altered regulation of channels and carriers is part of neoplastic transformation. Experimental modification of channel and transporter activity impacts tumour cell survival, proliferation, malignant progression, invasive behaviour or therapy resistance of tumour cells. A wide variety of distinct Ca(2+) permeable channels, K(+) channels, Na(+) channels and anion channels have been implicated in tumour growth and metastasis. Further experimental information is, however, needed to define the specific role of individual channel isoforms critically important for malignancy. Compelling experimental evidence supports the assumption that the pharmacological inhibition of ion channels or their regulators may be attractive targets to counteract tumour growth, prevent metastasis and overcome therapy resistance of tumour cells. This short review discusses the role of Ca(2+) permeable channels, K(+) channels, Na(+) channels and anion channels in tumour growth and metastasis and the therapeutic potential of respective inhibitors.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tuebingen, , Gmelinstrasse 5, Tübingen 72076, Germany
| | | |
Collapse
|
45
|
Turner KL, Sontheimer H. Cl- and K+ channels and their role in primary brain tumour biology. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130095. [PMID: 24493743 DOI: 10.1098/rstb.2013.0095] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Profound cell volume changes occur in primary brain tumours as they proliferate, invade surrounding tissue or undergo apoptosis. These volume changes are regulated by the flux of Cl(-) and K(+) ions and concomitant movement of water across the membrane, making ion channels pivotal to tumour biology. We discuss which specific Cl(-) and K(+) channels are involved in defined aspects of glioma biology and how these channels are regulated. Cl(-) is accumulated to unusually high concentrations in gliomas by the activity of the NKCC1 transporter and serves as an osmolyte and energetic driving force for volume changes. Cell volume condensation is required as cells enter M phase of the cell cycle and this pre-mitotic condensation is caused by channel-mediated ion efflux. Similarly, Cl(-) and K(+) channels dynamically regulate volume in invading glioma cells allowing them to adjust to small extracellular brain spaces. Finally, cell condensation is a hallmark of apoptosis and requires the concerted activation of Cl(-) and Ca(2+)-activated K(+) channels. Given the frequency of mutation and high importance of ion channels in tumour biology, the opportunity exists to target them for treatment.
Collapse
Affiliation(s)
- Kathryn L Turner
- Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, , 1719 6th Avenue South, CIRC 410, Birmingham, AL 35294, USA
| | | |
Collapse
|
46
|
Memmel S, Sukhorukov VL, Höring M, Westerling K, Fiedler V, Katzer A, Krohne G, Flentje M, Djuzenova CS. Cell surface area and membrane folding in glioblastoma cell lines differing in PTEN and p53 status. PLoS One 2014; 9:e87052. [PMID: 24498019 PMCID: PMC3909012 DOI: 10.1371/journal.pone.0087052] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/17/2013] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma multiforme (GBM) is characterized by rapid growth, invasion and resistance to chemo−/radiotherapy. The complex cell surface morphology with abundant membrane folds, microvilli, filopodia and other membrane extensions is believed to contribute to the highly invasive behavior and therapy resistance of GBM cells. The present study addresses the mechanisms leading to the excessive cell membrane area in five GBM lines differing in mutational status for PTEN and p53. In addition to scanning electron microscopy (SEM), the membrane area and folding were quantified by dielectric measurements of membrane capacitance using the single-cell electrorotation (ROT) technique. The osmotic stability and volume regulation of GBM cells were analyzed by video microscopy. The expression of PTEN, p53, mTOR and several other marker proteins involved in cell growth and membrane synthesis were examined by Western blotting. The combined SEM, ROT and osmotic data provided independent lines of evidence for a large variability in membrane area and folding among tested GBM lines. Thus, DK-MG cells (wild type p53 and wild type PTEN) exhibited the lowest degree of membrane folding, probed by the area-specific capacitance Cm = 1.9 µF/cm2. In contrast, cell lines carrying mutations in both p53 and PTEN (U373-MG and SNB19) showed the highest Cm values of 3.7–4.0 µF/cm2, which corroborate well with their heavily villated cell surface revealed by SEM. Since PTEN and p53 are well-known inhibitors of mTOR, the increased membrane area/folding in mutant GBM lines may be related to the enhanced protein and lipid synthesis due to a deregulation of the mTOR-dependent downstream signaling pathway. Given that membrane folds and extensions are implicated in tumor cell motility and metastasis, the dielectric approach presented here provides a rapid and simple tool for screening the biophysical cell properties in studies on targeting chemo- or radiotherapeutically the migration and invasion of GBM and other tumor types.
Collapse
Affiliation(s)
- Simon Memmel
- Lehrstuhl für Biotechnologie und Biophysik, Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Vladimir L. Sukhorukov
- Lehrstuhl für Biotechnologie und Biophysik, Universität Würzburg, Am Hubland, Würzburg, Germany
- * E-mail: (VLS); (CSD)
| | - Marcus Höring
- Lehrstuhl für Biotechnologie und Biophysik, Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Katherine Westerling
- Lehrstuhl für Biotechnologie und Biophysik, Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Vanessa Fiedler
- Department of Radiation Oncology, University Hospital Würzburg, Würzburg, Germany
| | - Astrid Katzer
- Department of Radiation Oncology, University Hospital Würzburg, Würzburg, Germany
| | - Georg Krohne
- Elektronenmikroskopie, Biozentrum, Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital Würzburg, Würzburg, Germany
| | - Cholpon S. Djuzenova
- Department of Radiation Oncology, University Hospital Würzburg, Würzburg, Germany
- * E-mail: (VLS); (CSD)
| |
Collapse
|
47
|
Catacuzzeno L, Michelucci A, Sforna L, Aiello F, Sciaccaluga M, Fioretti B, Castigli E, Franciolini F. Identification of key signaling molecules involved in the activation of the swelling-activated chloride current in human glioblastoma cells. J Membr Biol 2013; 247:45-55. [PMID: 24240542 DOI: 10.1007/s00232-013-9609-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/14/2013] [Indexed: 12/17/2022]
Abstract
The swelling-activated chloride current (I Cl,Vol) is abundantly expressed in glioblastoma (GBM) cells, where it controls cell volume and invasive migration. The transduction pathway mediating I Cl,Vol activation in GBM cells is, however, poorly understood. By means of pharmacological and electrophysiological approaches, on GL-15 human GBM cells we found that I Cl,Vol activation by hypotonic swelling required the activity of a U73122-sensitive phospholipase C (PLC). I Cl,Vol activation could also be induced by the membrane-permeable diacylglycerol (DAG) analog OAG. In contrast, neither calcium (Ca(2+)) chelation by BAPTA-AM nor changes in PKC activity were able to affect I Cl,Vol activation by hypotonic swelling. We further found that R59022, an inhibitor of diacylglycerol kinase (DGK), reverted I Cl,Vol activation, suggesting the involvement of phosphatidic acid. In addition, I Cl,Vol activation required the activity of a EHT1864-sensitive Rac1 small GTPase and the resulting actin polymerization, as I Cl,Vol activation was prevented by cytochalasin B. We finally show that I Cl,Vol can be activated by the promigratory fetal calf serum in a PLC- and DGK-dependent manner. This observation is potentially relevant because blood serum can likely come in contact with glioblastoma cells in vivo as a result of the tumor-related partial breakdown of the blood-brain barrier. Given the relevance of I Cl,Vol in GBM cell volume regulation and invasiveness, the several key signaling molecules found in this study to be involved in the activation of the I Cl,Vol may represent potential therapeutic targets against this lethal cancer.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Dipartimento di Biologia Cellulare e Ambientale, Universita' di Perugia, Via Pascoli 1, 06123, Perugia, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Williams S, Bateman A, O'Kelly I. Altered expression of two-pore domain potassium (K2P) channels in cancer. PLoS One 2013; 8:e74589. [PMID: 24116006 PMCID: PMC3792113 DOI: 10.1371/journal.pone.0074589] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/03/2013] [Indexed: 01/31/2023] Open
Abstract
Potassium channels have become a focus in cancer biology as they play roles in cell behaviours associated with cancer progression, including proliferation, migration and apoptosis. Two-pore domain (K2P) potassium channels are background channels which enable the leak of potassium ions from cells. As these channels are open at rest they have a profound effect on cellular membrane potential and subsequently the electrical activity and behaviour of cells in which they are expressed. The K2P family of channels has 15 mammalian members and already 4 members of this family (K2P2.1, K2P3.1, K2P9.1, K2P5.1) have been implicated in cancer. Here we examine the expression of all 15 members of the K2P family of channels in a range of cancer types. This was achieved using the online cancer microarray database, Oncomine (www.oncomine.org). Each gene was examined across 20 cancer types, comparing mRNA expression in cancer to normal tissue. This analysis revealed all but 3 K2P family members (K2P4.1, K2P16.1, K2P18.1) show altered expression in cancer. Overexpression of K2P channels was observed in a range of cancers including breast, leukaemia and lung while more cancers (brain, colorectal, gastrointestinal, kidney, lung, melanoma, oesophageal) showed underexpression of one or more channels. K2P1.1, K2P3.1, K2P12.1, were overexpressed in a range of cancers. While K2P1.1, K2P3.1, K2P5.1, K2P6.1, K2P7.1 and K2P10.1 showed significant underexpression across the cancer types examined. This analysis supports the view that specific K2P channels may play a role in cancer biology. Their altered expression together with their ability to impact the function of other ion channels and their sensitivity to environmental stimuli (pO2, pH, glucose, stretch) makes understanding the role these channels play in cancer of key importance.
Collapse
Affiliation(s)
- Sarah Williams
- Human Development and Health, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew Bateman
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ita O'Kelly
- Human Development and Health, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- * E-mail: I.M.O'
| |
Collapse
|
49
|
D'Alessandro G, Catalano M, Sciaccaluga M, Chece G, Cipriani R, Rosito M, Grimaldi A, Lauro C, Cantore G, Santoro A, Fioretti B, Franciolini F, Wulff H, Limatola C. KCa3.1 channels are involved in the infiltrative behavior of glioblastoma in vivo. Cell Death Dis 2013; 4:e773. [PMID: 23949222 PMCID: PMC3763441 DOI: 10.1038/cddis.2013.279] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/25/2013] [Accepted: 07/02/2013] [Indexed: 01/03/2023]
Abstract
Glioblastoma multiforme (GBM) is a diffuse brain tumor characterized by high infiltration in the brain parenchyma rendering the tumor difficult to eradicate by neurosurgery. Efforts to identify molecular targets involved in the invasive behavior of GBM suggested ion channel inhibition as a promising therapeutic approach. To determine if the Ca(2+)-dependent K(+) channel KCa3.1 could represent a key element for GBM brain infiltration, human GL-15 cells were xenografted into the brain of SCID mice that were then treated with the specific KCa3.1 blocker TRAM-34 (1-((2-chlorophenyl) (diphenyl)methyl)-1H-pyrazole). After 5 weeks of treatment, immunofluorescence analyses of cerebral slices revealed reduced tumor infiltration and astrogliosis surrounding the tumor, compared with untreated mice. Significant reduction of tumor infiltration was also observed in the brain of mice transplanted with KCa3.1-silenced GL-15 cells, indicating a direct effect of TRAM-34 on GBM-expressed KCa3.1 channels. As KCa3.1 channels are also expressed on microglia, we investigated the effects of TRAM-34 on microglia activation in GL-15 transplanted mice and found a reduction of CD68 staining in treated mice. Similar results were observed in vitro where TRAM-34 reduced both phagocytosis and chemotactic activity of primary microglia exposed to GBM-conditioned medium. Taken together, these results indicate that KCa3.1 activity has an important role in GBM invasiveness in vivo and that its inhibition directly affects glioma cell migration and reduces astrocytosis and microglia activation in response to tumor-released factors. KCa3.1 channel inhibition therefore constitutes a potential novel therapeutic approach to reduce GBM spreading into the surrounding tissue.
Collapse
Affiliation(s)
- G D'Alessandro
- Institute Pasteur, Cenci Bolognetti Foundation, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Huber SM, Butz L, Stegen B, Klumpp D, Braun N, Ruth P, Eckert F. Ionizing radiation, ion transports, and radioresistance of cancer cells. Front Physiol 2013; 4:212. [PMID: 23966948 PMCID: PMC3743404 DOI: 10.3389/fphys.2013.00212] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/24/2013] [Indexed: 12/22/2022] Open
Abstract
The standard treatment of many tumor entities comprises fractionated radiation therapy which applies ionizing radiation to the tumor-bearing target volume. Ionizing radiation causes double-strand breaks in the DNA backbone that result in cell death if the number of DNA double-strand breaks exceeds the DNA repair capacity of the tumor cell. Ionizing radiation reportedly does not only act on the DNA in the nucleus but also on the plasma membrane. In particular, ionizing radiation-induced modifications of ion channels and transporters have been reported. Importantly, these altered transports seem to contribute to the survival of the irradiated tumor cells. The present review article summarizes our current knowledge on the underlying mechanisms and introduces strategies to radiosensitize tumor cells by targeting plasma membrane ion transports.
Collapse
Affiliation(s)
- Stephan M Huber
- Department of Radiation Oncology, University of Tübingen Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|