1
|
Shrivastava A, Kumar A, Aggarwal LM, Pradhan S, Choudhary S, Ashish A, Kashyap K, Mishra S. Evolution of Bioelectric Membrane Potentials: Implications in Cancer Pathogenesis and Therapeutic Strategies. J Membr Biol 2024; 257:281-305. [PMID: 39183198 DOI: 10.1007/s00232-024-00323-2] [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: 05/30/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Electrophysiology typically deals with the electrical properties of excitable cells like neurons and muscles. However, all other cells (non-excitable) also possess bioelectric membrane potentials for intracellular and extracellular communications. These membrane potentials are generated by different ions present in fluids available in and outside the cell, playing a vital role in communication and coordination between the cell and its organelles. Bioelectric membrane potential variations disturb cellular ionic homeostasis and are characteristic of many diseases, including cancers. A rapidly increasing interest has emerged in sorting out the electrophysiology of cancer cells. Compared to healthy cells, the distinct electrical properties exhibited by cancer cells offer a unique way of understanding cancer development, migration, and progression. Decoding the altered bioelectric signals influenced by fluctuating electric fields benefits understanding cancer more closely. While cancer research has predominantly focussed on genetic and molecular traits, the delicate area of electrophysiological characteristics has increasingly gained prominence. This review explores the historical exploration of electrophysiology in the context of cancer cells, shedding light on how alterations in bioelectric membrane potentials, mediated by ion channels and gap junctions, contribute to the pathophysiology of cancer.
Collapse
Affiliation(s)
- Anju Shrivastava
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India.
| | - Amit Kumar
- Department of Anatomy, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Lalit Mohan Aggarwal
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Satyajit Pradhan
- Radiation Oncology, Mahamana Pandit Madhan Mohan Malaviya Cancer Centre, Varanasi, India
| | - Sunil Choudhary
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Keshav Kashyap
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Shivani Mishra
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
2
|
Kofman K, Levin M. Bioelectric pharmacology of cancer: A systematic review of ion channel drugs affecting the cancer phenotype. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 191:25-39. [PMID: 38971325 DOI: 10.1016/j.pbiomolbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
Cancer is a pernicious and pressing medical problem; moreover, it is a failure of multicellular morphogenesis that sheds much light on evolutionary developmental biology. Numerous classes of pharmacological agents have been considered as cancer therapeutics and evaluated as potential carcinogenic agents; however, these are spread throughout the primary literature. Here, we briefly review recent work on ion channel drugs as promising anti-cancer treatments and present a systematic review of the known cancer-relevant effects of 109 drugs targeting ion channels. The roles of ion channels in cancer are consistent with the importance of bioelectrical parameters in cell regulation and with the functions of bioelectric signaling in morphogenetic signals that act as cancer suppressors. We find that compounds that are well-known for having targets in the nervous system, such as voltage-gated ion channels, ligand-gated ion channels, proton pumps, and gap junctions are especially relevant to cancer. Our review suggests further opportunities for the repurposing of numerous promising candidates in the field of cancer electroceuticals.
Collapse
Affiliation(s)
- Karina Kofman
- Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Michael Levin
- Allen Discovery Center at Tufts University, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, USA.
| |
Collapse
|
3
|
Min Q, Gao Y, Wang Y. Bioelectricity in dental medicine: a narrative review. Biomed Eng Online 2024; 23:3. [PMID: 38172866 PMCID: PMC10765628 DOI: 10.1186/s12938-023-01189-6] [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: 09/07/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Bioelectric signals, whether exogenous or endogenous, play crucial roles in the life processes of organisms. Recently, the significance of bioelectricity in the field of dentistry is steadily gaining greater attention. OBJECTIVE This narrative review aims to comprehensively outline the theory, physiological effects, and practical applications of bioelectricity in dental medicine and to offer insights into its potential future direction. It attempts to provide dental clinicians and researchers with an electrophysiological perspective to enhance their clinical practice or fundamental research endeavors. METHODS An online computer search for relevant literature was performed in PubMed, Web of Science and Cochrane Library, with the keywords "bioelectricity, endogenous electric signal, electric stimulation, dental medicine." RESULTS Eventually, 288 documents were included for review. The variance in ion concentration between the interior and exterior of the cell membrane, referred to as transmembrane potential, forms the fundamental basis of bioelectricity. Transmembrane potential has been established as an essential regulator of intercellular communication, mechanotransduction, migration, proliferation, and immune responses. Thus, exogenous electric stimulation can significantly alter cellular action by affecting transmembrane potential. In the field of dental medicine, electric stimulation has proven useful for assessing pulp condition, locating root apices, improving the properties of dental biomaterials, expediting orthodontic tooth movement, facilitating implant osteointegration, addressing maxillofacial malignancies, and managing neuromuscular dysfunction. Furthermore, the reprogramming of bioelectric signals holds promise as a means to guide organism development and intervene in disease processes. Besides, the development of high-throughput electrophysiological tools will be imperative for identifying ion channel targets and precisely modulating bioelectricity in the future. CONCLUSIONS Bioelectricity has found application in various concepts of dental medicine but large-scale, standardized, randomized controlled clinical trials are still necessary in the future. In addition, the precise, repeatable and predictable measurement and modulation methods of bioelectric signal patterns are essential research direction.
Collapse
Affiliation(s)
- Qingqing Min
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, 214000, China
| | - Yajun Gao
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, 214000, China
| | - Yao Wang
- Department of Implantology, Wuxi Stomatology Hospital, Wuxi, 214000, China.
| |
Collapse
|
4
|
Fontani V, Cruciani S, Santaniello S, Rinaldi S, Maioli M. Impact of REAC Regenerative Endogenous Bioelectrical Cell Reprogramming on MCF7 Breast Cancer Cells. J Pers Med 2023; 13:1019. [PMID: 37374009 DOI: 10.3390/jpm13061019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/07/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Human breast adenocarcinoma is a form of cancer which has the tendency to metastasize to other tissues, including bones, lungs, brain, and liver. Several chemotherapeutic drugs are used to treat breast tumors. Their combination is used to simultaneously target different mechanisms involved in cell replication. Radio electric asymmetric conveyer (REAC) technology is an innovative technology, used both in vitro and in vivo, to induce cell reprogramming and counteract senescence processes. Within this context, we treated MCF-7 cells with a regenerative (RGN) REAC treatment for a period ranging between 3 and 7 days. We then analyzed cell viability by trypan blue assays and gene and protein expression by real time-qPCR and confocal microscope, respectively. We also detected the levels of the main proteins involved in tumor progression, DKK1 and SFRP1, by ELISA and cell senescence by β-galactosidase tests. Our results showed the ability of REAC RGN to counteract MCF-7 proliferation, probably inducing autophagy via the upregulation of Beclin-1 and LC3-I, and the modulation of specific tumorigenic biomarkers, such as DKK1 and SPFR1. Our results could suggest the application of the REAC RGN in future in vivo experiments, as an aid for the therapeutic strategies usually applied for breast cancer treatment.
Collapse
Affiliation(s)
- Vania Fontani
- Department of Regenerative Medicine, Rinaldi Fontani Institute, 50144 Florence, Italy
- Department of Adaptive Neuro Psycho Physio Pathology and Neuro Psycho Physical Optimization, Rinaldi Fontani Institute, 50144 Florence, Italy
- Research Department, Rinaldi Fontani Foundation, 50144 Florence, Italy
| | - Sara Cruciani
- Research Department, Rinaldi Fontani Foundation, 50144 Florence, Italy
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Sara Santaniello
- Research Department, Rinaldi Fontani Foundation, 50144 Florence, Italy
| | - Salvatore Rinaldi
- Department of Regenerative Medicine, Rinaldi Fontani Institute, 50144 Florence, Italy
- Department of Adaptive Neuro Psycho Physio Pathology and Neuro Psycho Physical Optimization, Rinaldi Fontani Institute, 50144 Florence, Italy
- Research Department, Rinaldi Fontani Foundation, 50144 Florence, Italy
| | - Margherita Maioli
- Research Department, Rinaldi Fontani Foundation, 50144 Florence, Italy
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
5
|
Pio-Lopez L, Levin M. Morphoceuticals: perspectives for discovery of drugs targeting anatomical control mechanisms in regenerative medicine, cancer and aging. Drug Discov Today 2023; 28:103585. [PMID: 37059328 DOI: 10.1016/j.drudis.2023.103585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/18/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Morphoceuticals are a new class of interventions that target the setpoints of anatomical homeostasis for efficient, modular control of growth and form. Here, we focus on a subclass: electroceuticals, which specifically target the cellular bioelectrical interface. Cellular collectives in all tissues form bioelectrical networks via ion channels and gap junctions that process morphogenetic information, controlling gene expression and allowing cell networks to adaptively and dynamically control growth and pattern formation. Recent progress in understanding this physiological control system, including predictive computational models, suggests that targeting bioelectrical interfaces can control embryogenesis and maintain shape against injury, senescence and tumorigenesis. We propose a roadmap for drug discovery focused on manipulating endogenous bioelectric signaling for regenerative medicine, cancer suppression and antiaging therapeutics. Teaser: By taking advantage of the native problem-solving competencies of cells and tissues, a new kind of top-down approach to biomedicine becomes possible. Bioelectricity offers an especially tractable interface for interventions targeting the software of life for regenerative medicine applications.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
Huang X, Tanveer M, Min Y, Shabala S. Melatonin as a regulator of plant ionic homeostasis: implications for abiotic stress tolerance. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:5886-5902. [PMID: 35640481 DOI: 10.1093/jxb/erac224] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/23/2022] [Indexed: 06/15/2023]
Abstract
Melatonin is a highly conserved and ubiquitous molecule that operates upstream of a broad array of receptors in animal systems. Since melatonin was discovered in plants in 1995, hundreds of papers have been published revealing its role in plant growth, development, and adaptive responses to the environment. This paper summarizes the current state of knowledge of melatonin's involvement in regulating plant ion homeostasis and abiotic stress tolerance. The major topics covered here are: (i) melatonin's control of H+-ATPase activity and its implication for plant adaptive responses to various abiotic stresses; (ii) regulation of the reactive oxygen species (ROS)-Ca2+ hub by melatonin and its role in stress signaling; and (iii) melatonin's regulation of ionic homeostasis via hormonal cross-talk. We also show that the properties of the melatonin molecule allow its direct scavenging of ROS, thus preventing negative effects of ROS-induced activation of ion channels. The above 'desensitization' may play a critical role in preventing stress-induced K+ loss from the cytosol as well as maintaining basic levels of cytosolic Ca2+ required for optimal cell operation. Future studies should focus on revealing the molecular identity of transporters that could be directly regulated by melatonin and providing a bioinformatic analysis of evolutionary aspects of melatonin sensing and signaling.
Collapse
Affiliation(s)
- Xin Huang
- International Research Center for Environmental Membrane Biology, Foshan University, Foshan, Guangdong, China
| | - Mohsin Tanveer
- Tasmanian Institute of Agriculture, University of Tasmania, Tas, Hobart, Australia
| | - Yu Min
- International Research Center for Environmental Membrane Biology, Foshan University, Foshan, Guangdong, China
| | - Sergey Shabala
- International Research Center for Environmental Membrane Biology, Foshan University, Foshan, Guangdong, China
- Tasmanian Institute of Agriculture, University of Tasmania, Tas, Hobart, Australia
- School of Biological Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
7
|
Pai VP, Cooper BG, Levin M. Screening Biophysical Sensors and Neurite Outgrowth Actuators in Human Induced-Pluripotent-Stem-Cell-Derived Neurons. Cells 2022; 11:cells11162470. [PMID: 36010547 PMCID: PMC9406775 DOI: 10.3390/cells11162470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
All living cells maintain a charge distribution across their cell membrane (membrane potential) by carefully controlled ion fluxes. These bioelectric signals regulate cell behavior (such as migration, proliferation, differentiation) as well as higher-level tissue and organ patterning. Thus, voltage gradients represent an important parameter for diagnostics as well as a promising target for therapeutic interventions in birth defects, injury, and cancer. However, despite much progress in cell and molecular biology, little is known about bioelectric states in human stem cells. Here, we present simple methods to simultaneously track ion dynamics, membrane voltage, cell morphology, and cell activity (pH and ROS), using fluorescent reporter dyes in living human neurons derived from induced neural stem cells (hiNSC). We developed and tested functional protocols for manipulating ion fluxes, membrane potential, and cell activity, and tracking neural responses to injury and reinnervation in vitro. Finally, using morphology sensor, we tested and quantified the ability of physiological actuators (neurotransmitters and pH) to manipulate nerve repair and reinnervation. These methods are not specific to a particular cell type and should be broadly applicable to the study of bioelectrical controls across a wide range of combinations of models and endpoints.
Collapse
Affiliation(s)
- Vaibhav P. Pai
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Ben G. Cooper
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
- Correspondence:
| |
Collapse
|
8
|
Tassinari R, Cavallini C, Olivi E, Facchin F, Taglioli V, Zannini C, Marcuzzi M, Ventura C. Cell Responsiveness to Physical Energies: Paving the Way to Decipher a Morphogenetic Code. Int J Mol Sci 2022; 23:ijms23063157. [PMID: 35328576 PMCID: PMC8949133 DOI: 10.3390/ijms23063157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
We discuss emerging views on the complexity of signals controlling the onset of biological shapes and functions, from the nanoarchitectonics arising from supramolecular interactions, to the cellular/multicellular tissue level, and up to the unfolding of complex anatomy. We highlight the fundamental role of physical forces in cellular decisions, stressing the intriguing similarities in early morphogenesis, tissue regeneration, and oncogenic drift. Compelling evidence is presented, showing that biological patterns are strongly embedded in the vibrational nature of the physical energies that permeate the entire universe. We describe biological dynamics as informational processes at which physics and chemistry converge, with nanomechanical motions, and electromagnetic waves, including light, forming an ensemble of vibrations, acting as a sort of control software for molecular patterning. Biomolecular recognition is approached within the establishment of coherent synchronizations among signaling players, whose physical nature can be equated to oscillators tending to the coherent synchronization of their vibrational modes. Cytoskeletal elements are now emerging as senders and receivers of physical signals, "shaping" biological identity from the cellular to the tissue/organ levels. We finally discuss the perspective of exploiting the diffusive features of physical energies to afford in situ stem/somatic cell reprogramming, and tissue regeneration, without stem cell transplantation.
Collapse
Affiliation(s)
- Riccardo Tassinari
- ELDOR LAB, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Via Gobetti 101, 40129 Bologna, Italy; (R.T.); (C.C.); (E.O.); (V.T.); (C.Z.)
| | - Claudia Cavallini
- ELDOR LAB, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Via Gobetti 101, 40129 Bologna, Italy; (R.T.); (C.C.); (E.O.); (V.T.); (C.Z.)
| | - Elena Olivi
- ELDOR LAB, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Via Gobetti 101, 40129 Bologna, Italy; (R.T.); (C.C.); (E.O.); (V.T.); (C.Z.)
| | - Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Valentina Taglioli
- ELDOR LAB, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Via Gobetti 101, 40129 Bologna, Italy; (R.T.); (C.C.); (E.O.); (V.T.); (C.Z.)
| | - Chiara Zannini
- ELDOR LAB, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Via Gobetti 101, 40129 Bologna, Italy; (R.T.); (C.C.); (E.O.); (V.T.); (C.Z.)
| | - Martina Marcuzzi
- INBB, Biostructures and Biosystems National Institute, Viale Medaglie d’Oro 305, 00136 Rome, Italy;
| | - Carlo Ventura
- ELDOR LAB, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Via Gobetti 101, 40129 Bologna, Italy; (R.T.); (C.C.); (E.O.); (V.T.); (C.Z.)
- Correspondence: ; Tel.: +39-347-920-6992
| |
Collapse
|
9
|
Ion Channel Drugs Suppress Cancer Phenotype in NG108-15 and U87 Cells: Toward Novel Electroceuticals for Glioblastoma. Cancers (Basel) 2022; 14:cancers14061499. [PMID: 35326650 PMCID: PMC8946312 DOI: 10.3390/cancers14061499] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma is a lethal brain cancer that commonly recurs after tumor resection and chemotherapy treatment. Depolarized resting membrane potentials and an acidic intertumoral extracellular pH have been associated with a proliferative state and drug resistance, suggesting that forced hyperpolarization and disruption of proton pumps in the plasma membrane could be a successful strategy for targeting glioblastoma overgrowth. We screened 47 compounds and compound combinations, most of which were ion-modulating, at different concentrations in the NG108-15 rodent neuroblastoma/glioma cell line. A subset of these were tested in the U87 human glioblastoma cell line. A FUCCI cell cycle reporter was stably integrated into both cell lines to monitor proliferation and cell cycle response. Immunocytochemistry, electrophysiology, and a panel of physiological dyes reporting voltage, calcium, and pH were used to characterize responses. The most effective treatments on proliferation in U87 cells were combinations of NS1643 and pantoprazole; retigabine and pantoprazole; and pantoprazole or NS1643 with temozolomide. Marker analysis and physiological dye signatures suggest that exposure to bioelectric drugs significantly reduces proliferation, makes the cells senescent, and promotes differentiation. These results, along with the observed low toxicity in human neurons, show the high efficacy of electroceuticals utilizing combinations of repurposed FDA approved drugs.
Collapse
|
10
|
Hao P, Song KY, Wang SQ, Huang XJ, Yao DW, Yang DJ. ABCC9 Is Downregulated and Prone to Microsatellite Instability on ABCC9tetra in Canine Breast Cancer. Front Vet Sci 2022; 8:819293. [PMID: 35071399 PMCID: PMC8777218 DOI: 10.3389/fvets.2021.819293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Tumorigenesis is associated with metabolic abnormalities and genomic instability. Microsatellite mutations, including microsatellite instability (MSI) and loss of heterozygosity (LOH), are associated with the functional impairment of some tumor-related genes. To investigate the role of MSI and LOH in sporadic breast tumors in canines, 22 tumors DNA samples and their adjacent normal tissues were evaluated using polyacrylamide gel electrophoresis and silver staining for 58 microsatellites. Quantitative real-time polymerase chain reaction, promoter methylation analysis and immunohistochemical staining were used to quantify gene expression. The results revealed that a total of 14 tumors (6 benign tumors and 8 breast cancers) exhibited instability as MSI-Low tumors. Most of the microsatellite loci possessed a single occurrence of mutations. The maximum number of MSI mutations on loci was observed in tumors with a lower degree of differentiation. Among the unstable markers, FH2060 (4/22), ABCC9tetra (4/22) and SCN11A (6/22) were high-frequency mutation sites, whereas FH2060 was a high-frequency LOH site (4/22). The ABCC9tetra locus was mutated only in cancerous tissue, although it was excluded by transcription. The corresponding genes and proteins were significantly downregulated in malignant tissues, particularly in tumors with MSI. Furthermore, the promoter methylation results of the adenosine triphosphate binding cassette subfamily C member 9 (ABCC9) showed that there was a high level of methylation in breast tissues, but only one case showed a significant elevation compared with the control. In conclusion, MSI-Low or MSI-Stable is characteristic of most sporadic mammary tumors. Genes associated with tumorigenesis are more likely to develop MSI. ABCC9 protein and transcription abnormalities may be associated with ABCC9tetra instability.
Collapse
Affiliation(s)
- Pan Hao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kai-Yue Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Si-Qi Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiao-Jun Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Da-Wei Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - De-Ji Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
11
|
Payne SL, Ram P, Srinivasan DH, Le TT, Levin M, Oudin MJ. Potassium channel-driven bioelectric signalling regulates metastasis in triple-negative breast cancer. EBioMedicine 2022; 75:103767. [PMID: 34933180 PMCID: PMC8688589 DOI: 10.1016/j.ebiom.2021.103767] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There is a critical need to better understand the mechanisms that drive local cell invasion and metastasis to develop new therapeutics targeting metastatic disease. Bioelectricity is an important mediator of cellular processes and changes in the resting membrane potential (RMP) are associated with increased cancer cell invasion. However, whether the RMP can be used to target invading cancer cells is unknown. METHODS We employed both genetic and pharmacological manipulation of potassium channel activity and characterized the effects on breast cancer cell migration and invasion in vitro, and metastasis in an animal model of breast cancer. FINDINGS Our data demonstrate that altering the RMP of triple-negative breast cancer (TNBC) cells by manipulating potassium channel expression increases in vitro invasion, in vivo tumour growth and metastasis, and is accompanied by changes in gene expression associated with cell adhesion. INTERPRETATION We describe a novel mechanism for RMP-mediated cell migration involving cadherin-11 and the MAPK pathway. Importantly, we identify a new strategy to target metastatic TNBC in vivo by repurposing an FDA-approved potassium channel blocker. Our results demonstrate that bioelectricity regulates cancer cell invasion and metastasis which could lead to a new class of therapeutics for patients with metastatic disease. FUNDING This work was supported by the National Institutes of Health (R00-CA207866 to M.J.O.), Tufts University (Start-up funds from the School of Engineering to M.J.O., Tufts Collaborates Award to M.J.O. and M.L.), Allen Discovery centre program (Paul G. Allen Frontiers Group (12,171) to M.L.), and Breast Cancer Alliance Young Investigator Grant to M.J.O, Laidlaw Scholar funding to D.S. M.L. also gratefully acknowledges support of the Barton Family Foundation.
Collapse
Affiliation(s)
- Samantha L Payne
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, United States of America
| | - Priyanka Ram
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, United States of America
| | - Deepti H Srinivasan
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, United States of America
| | - Thanh T Le
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, United States of America
| | - Michael Levin
- Allen Discovery Center, 200 College Avenue, Tufts University, Medford, MA 02155, United States of America
| | - Madeleine J Oudin
- Department of Biomedical Engineering, 200 College Avenue, Tufts University, Medford MA 02155, United States of America.
| |
Collapse
|
12
|
Tassinari R, Cavallini C, Olivi E, Taglioli V, Zannini C, Ventura C. Unveiling the morphogenetic code: A new path at the intersection of physical energies and chemical signaling. World J Stem Cells 2021; 13:1382-1393. [PMID: 34786150 PMCID: PMC8567452 DOI: 10.4252/wjsc.v13.i10.1382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/16/2021] [Accepted: 09/10/2021] [Indexed: 02/06/2023] Open
Abstract
In this editorial, we discuss the remarkable role of physical energies in the control of cell signaling networks and in the specification of the architectural plan of both somatic and stem cells. In particular, we focus on the biological relevance of bioelectricity in the pattern control that orchestrates both developmental and regenerative pathways. To this end, the narrative starts from the dawn of the first studies on animal electricity, reconsidering the pioneer work of Harold Saxton Burr in the light of the current achievements. We finally discuss the most recent evidence showing that bioelectric signaling is an essential component of the informational processes that control pattern specification during embryogenesis, regeneration, or even malignant transformation. We conclude that there is now mounting evidence for the existence of a Morphogenetic Code, and that deciphering this code may lead to unprecedented opportunities for the development of novel paradigms of cure in regenerative and precision medicine.
Collapse
Affiliation(s)
- Riccardo Tassinari
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – ELDOR LAB, Bologna 40129, Italy
| | - Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – ELDOR LAB, Bologna 40129, Italy
| | - Elena Olivi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – ELDOR LAB, Bologna 40129, Italy
| | - Valentina Taglioli
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – ELDOR LAB, Bologna 40129, Italy
| | - Chiara Zannini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – ELDOR LAB, Bologna 40129, Italy
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – ELDOR LAB, Bologna 40129, Italy
| |
Collapse
|
13
|
Cell Systems Bioelectricity: How Different Intercellular Gap Junctions Could Regionalize a Multicellular Aggregate. Cancers (Basel) 2021; 13:cancers13215300. [PMID: 34771463 PMCID: PMC8582473 DOI: 10.3390/cancers13215300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 01/10/2023] Open
Abstract
Electric potential distributions can act as instructive pre-patterns for development, regeneration, and tumorigenesis in cell systems. The biophysical states influence transcription, proliferation, cell shape, migration, and differentiation through biochemical and biomechanical downstream transduction processes. A major knowledge gap is the origin of spatial patterns in vivo, and their relationship to the ion channels and the electrical synapses known as gap junctions. Understanding this is critical for basic evolutionary developmental biology as well as for regenerative medicine. We computationally show that cells may express connexin proteins with different voltage-gated gap junction conductances as a way to maintain multicellular regions at distinct membrane potentials. We show that increasing the multicellular connectivity via enhanced junction function does not always contribute to the bioelectrical normalization of abnormally depolarized multicellular patches. From a purely electrical junction view, this result suggests that the reduction rather than the increase of specific connexin levels can also be a suitable bioelectrical approach in some cases and time stages. We offer a minimum model that incorporates effective conductances ultimately related to specific ion channel and junction proteins that are amenable to external regulation. We suggest that the bioelectrical patterns and their encoded instructive information can be externally modulated by acting on the mean fields of cell systems, a complementary approach to that of acting on the molecular characteristics of individual cells. We believe that despite the limitations of a biophysically focused model, our approach can offer useful qualitative insights into the collective dynamics of cell system bioelectricity.
Collapse
|
14
|
Bhattacharya D, Gawali VS, Kallay L, Toukam DK, Koehler A, Stambrook P, Krummel DP, Sengupta S. Therapeutically leveraging GABA A receptors in cancer. Exp Biol Med (Maywood) 2021; 246:2128-2135. [PMID: 34649481 DOI: 10.1177/15353702211032549] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
γ-aminobutyric acid or GABA is an amino acid that functionally acts as a neurotransmitter and is critical to neurotransmission. GABA is also a metabolite in the Krebs cycle. It is therefore unsurprising that GABA and its receptors are also present outside of the central nervous system, including in immune cells. This observation suggests that GABAergic signaling impacts events beyond brain function and possibly human health beyond neurological disorders. Indeed, GABA receptor subunits are expressed in pathological disease states, including in disparate cancers. The role that GABA and its receptors may play in cancer development and progression remains unclear. If, however, those cancers have functional GABA receptors that participate in GABAergic signaling, it raises an important question whether these signaling pathways might be targetable for therapeutic benefit. Herein we summarize the effects of modulating Type-A GABA receptor signaling in various cancers and highlight how Type-A GABA receptors could emerge as a novel therapeutic target in cancer.
Collapse
Affiliation(s)
- Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Vaibhavkumar S Gawali
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Laura Kallay
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Donatien K Toukam
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Abigail Koehler
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Peter Stambrook
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
15
|
Hu C, Zuo H, Li Y. Effects of Radiofrequency Electromagnetic Radiation on Neurotransmitters in the Brain. Front Public Health 2021; 9:691880. [PMID: 34485223 PMCID: PMC8415840 DOI: 10.3389/fpubh.2021.691880] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/13/2021] [Indexed: 12/29/2022] Open
Abstract
With the rapid development of electronic information in the past 30 years, technical achievements based on electromagnetism have been widely used in various fields pertaining to human production and life. Consequently, electromagnetic radiation (EMR) has become a substantial new pollution source in modern civilization. The biological effects of EMR have attracted considerable attention worldwide. The possible interaction of EMR with human organs, especially the brain, is currently where the most attention is focused. Many studies have shown that the nervous system is an important target organ system sensitive to EMR. In recent years, an increasing number of studies have focused on the neurobiological effects of EMR, including the metabolism and transport of neurotransmitters. As messengers of synaptic transmission, neurotransmitters play critical roles in cognitive and emotional behavior. Here, the effects of EMR on the metabolism and receptors of neurotransmitters in the brain are summarized.
Collapse
Affiliation(s)
- Cuicui Hu
- Anhui Medical University, Academy of Life Sciences, Hefei, China.,Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yang Li
- Anhui Medical University, Academy of Life Sciences, Hefei, China.,Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
16
|
Casey T, Suarez-Trujillo A, Cummings S, Huff K, Crodian J, Bhide K, Aduwari C, Teeple K, Shamay A, Mabjeesh SJ, San Miguel P, Thimmapuram J, Plaut K. Core circadian clock transcription factor BMAL1 regulates mammary epithelial cell growth, differentiation, and milk component synthesis. PLoS One 2021; 16:e0248199. [PMID: 34415905 PMCID: PMC8378744 DOI: 10.1371/journal.pone.0248199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/06/2021] [Indexed: 11/18/2022] Open
Abstract
The role the mammary epithelial circadian clock plays in gland development and lactation is unknown. We hypothesized that mammary epithelial clocks function to regulate mammogenesis and lactogenesis, and propose the core clock transcription factor BMAL1:CLOCK regulates genes that control mammary epithelial development and milk synthesis. Our objective was to identify transcriptional targets of BMAL1 in undifferentiated (UNDIFF) and lactogen differentiated (DIFF) mammary epithelial cells (HC11) using ChIP-seq. Ensembl gene IDs with the nearest transcriptional start site to ChIP-seq peaks were explored as potential targets, and represented 846 protein coding genes common to UNDIFF and DIFF cells and 2773 unique to DIFF samples. Genes with overlapping peaks between samples (1343) enriched cell-cell adhesion, membrane transporters and lipid metabolism categories. To functionally verify targets, an HC11 line with Bmal1 gene knocked out (BMAL1-KO) using CRISPR-CAS was created. BMAL1-KO cultures had lower cell densities over an eight-day growth curve, which was associated with increased (p<0.05) levels of reactive oxygen species and lower expression of superoxide dismutase 3 (Sod3). RT-qPCR analysis also found lower expression of the putative targets, prolactin receptor (Prlr), Ppara, and beta-casein (Csn2). Findings support our hypothesis and highlight potential importance of clock in mammary development and substrate transport.
Collapse
Affiliation(s)
- Theresa Casey
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Aridany Suarez-Trujillo
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Shelby Cummings
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Katelyn Huff
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Jennifer Crodian
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Ketaki Bhide
- Bioinformatics Core, Purdue University, West Lafayette, IN, United States of America
| | - Clare Aduwari
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Kelsey Teeple
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Avi Shamay
- Animal Science Institute, Agriculture Research Origination, The Volcani Center, Rishon Letsiyon, Israel
| | - Sameer J. Mabjeesh
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Phillip San Miguel
- Genomics Core, Purdue University, West Lafayette, IN, United States of America
| | - Jyothi Thimmapuram
- Bioinformatics Core, Purdue University, West Lafayette, IN, United States of America
| | - Karen Plaut
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
17
|
Mravec B. Neurobiology of Cancer: Introduction of New Drugs in the Treatment and Prevention of Cancer. Int J Mol Sci 2021; 22:6115. [PMID: 34204103 PMCID: PMC8201304 DOI: 10.3390/ijms22116115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
Research on the neurobiology of cancer, which lies at the border of neuroscience and oncology, has elucidated the mechanisms and pathways that enable the nervous system to modulate processes associated with cancer initiation and progression. This research has also shown that several drugs which modulate interactions between the nervous system and the tumor micro- and macroenvironments significantly reduced the progression of cancer in animal models. Encouraging results were also provided by prospective clinical trials investigating the effect of drugs that reduce adrenergic signaling on the course of cancer in oncological patients. Moreover, it has been shown that reducing adrenergic signaling might also reduce the incidence of cancer in animal models, as well as in humans. However, even if many experimental and clinical findings have confirmed the preventive and therapeutic potential of drugs that reduce the stimulatory effect of the nervous system on processes related to cancer initiation and progression, several questions remain unanswered. Therefore, the aim of this review is to critically evaluate the efficiency of these drugs and to discuss questions that need to be answered before their introduction into conventional cancer treatment and prevention.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 813 72 Bratislava, Slovakia; ; Tel.: +421-(2)-59357527; Fax: +421-(2)-59357601
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| |
Collapse
|
18
|
Phillips JA, Hutchings C, Djamgoz MBA. Clinical Potential of Nerve Input to Tumors: A Bioelectricity Perspective. Bioelectricity 2021; 3:14-26. [PMID: 34476375 DOI: 10.1089/bioe.2020.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
We support the notion that the neural connections of the tumor microenvironment (TME) and the associated 'bioelectricity' play significant role in the pathophysiology of cancer. In several cancers, the nerve input promotes the cancer process. While straightforward surgical denervation of tumors, therefore, could improve prognosis, resulting side effects of such a procedure would be unpredictable and irreversible. On the other hand, tumor innervation can be manipulated effectively for therapeutic purposes by alternative novel approaches broadly termed "electroceuticals." In this perspective, we evaluate the clinical potential of targeting the TME first through manipulation of the nerve input itself and second by application of electric fields directly to the tumor. The former encompasses several different biophysical and biochemical approaches. These include implantable devices, nanoparticles, and electroactive polymers, as well as optogenetics and chemogenetics. As regard bioelectrical manipulation of the tumor itself, the "tumor-treating field" technique, applied to gliomas commonly in combination with chemotherapy, is evaluated. Also, as electroceuticals, drugs acting on ion channels and neurotransmitter receptors are highlighted for completeness. It is concluded, first, that electroceuticals comprise a broad range of biomedical tools. Second, such electroceuticals present significant clinical potential for exploiting the neural component of the TME as a strategy against cancer. Finally, the inherent bioelectric characteristics of tumors themselves are also amenable to complementary approaches. Collectively, these represent an evolving, dynamic field and further progress and applications can be expected to follow both conceptually and technically.
Collapse
Affiliation(s)
- Jade A Phillips
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charlotte Hutchings
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Biotechnology Research Center, Cyprus International University, Nicosia, North Cyprus
| |
Collapse
|
19
|
Ion Channels in Cancer: Orchestrators of Electrical Signaling and Cellular Crosstalk. Rev Physiol Biochem Pharmacol 2020; 183:103-133. [PMID: 32894333 DOI: 10.1007/112_2020_48] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ion channels are pore-forming transmembrane proteins that govern ion flux to regulate a myriad of biological processes in development, physiology, and disease. Across various types of cancer, ion channel expression and activity are often dysregulated. We review the contribution of ion channels to multiple stages of tumorigenesis based on data from in vivo model systems. As intertumoral and intratumoral heterogeneities are major obstacles in developing effective therapies, we provide perspectives on how ion channels in tumor cells and their microenvironment represent targetable vulnerabilities in the areas of tumor-stromal cell interactions, cancer neuroscience, and cancer mechanobiology.
Collapse
|
20
|
Bhattacharya S, Hyland C, Falk MM, Iovine MK. Connexin 43 gap junctional intercellular communication inhibits evx1 expression and joint formation in regenerating fins. Development 2020; 147:dev.190512. [PMID: 32541014 DOI: 10.1242/dev.190512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/05/2020] [Indexed: 12/23/2022]
Abstract
The gap junction protein Connexin 43 (Cx43) contributes to cell fate decisions that determine the location of fin ray joints during regeneration. Here, we provide insights into how Cx43, expressed medially, influences changes in gene expression in lateral skeletal precursor cells. Using the Gap27 peptide inhibitor specific to Cx43, we show that Cx43-gap junctional intercellular communication (GJIC) influences Cx43-dependent skeletal phenotypes, including segment length. We also demonstrate that Cx43-GJIC influences the expression of the Smp/β-catenin pathway in the lateral skeletal precursor cells, and does not influence the Sema3d pathway. Moreover, we show that the cx43lh10 allele, which has increased Cx43 protein levels, exhibits increased regenerate length and segment length. These phenotypes are rescued by Gap27, suggesting that increased Cx43 is responsible for the observed Cx43 phenotypes. Finally, our findings suggest that inhibition of Cx43 hemichannel activity does not influence Cx43-dependent skeletal phenotypes. These data provide evidence that Cx43-GJIC is responsible for regulating cell fate decisions associated with appropriate joint formation in the regenerating fin.
Collapse
Affiliation(s)
| | - Caitlin Hyland
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18020, USA
| | - Matthias M Falk
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18020, USA
| | - M Kathryn Iovine
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18020, USA
| |
Collapse
|
21
|
Abstract
Selective serotonin reuptake inhibitor (SSRI) drugs, targeting serotonin transport, are widely used. A puzzling and biomedically important phenomenon concerns the persistent sexual dysfunction following SSRI use seen in some patients. What could be the mechanism of a persistent physiological state brought on by a transient exposure to serotonin transport blockers? In this study, we briefly review the clinical facts concerning this side effect of serotonin reuptake inhibitors and suggest a possible mechanism. Bioelectric circuits (among neural or non-neural cells) could persistently maintain alterations of bioelectric cell properties (resting potential), resulting in long-term changes in electrophysiology and signaling. We present new data revealing this phenomenon in planarian flatworms, in which brief SSRI exposures induce long-lasting changes in resting potential profile. We also briefly review recent data linking neurotransmitter signaling to developmental bioelectrics. Further study of tissue bioelectric memory could enable the design of ionoceutical interventions to counteract side effects of SSRIs and similar drugs.
Collapse
Affiliation(s)
- David Healy
- Hergest Unit, Department of Psychiatry, Bangor University, Bangor, Wales
| | - Joshua LaPalme
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| |
Collapse
|
22
|
Levin M, Selberg J, Rolandi M. Endogenous Bioelectrics in Development, Cancer, and Regeneration: Drugs and Bioelectronic Devices as Electroceuticals for Regenerative Medicine. iScience 2019; 22:519-533. [PMID: 31837520 PMCID: PMC6920204 DOI: 10.1016/j.isci.2019.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/15/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
A major frontier in the post-genomic era is the investigation of the control of coordinated growth and three-dimensional form. Dynamic remodeling of complex organs in regulative embryogenesis, regeneration, and cancer reveals that cells and tissues make decisions that implement complex anatomical outcomes. It is now essential to understand not only the genetics that specifies cellular hardware but also the physiological software that implements tissue-level plasticity and robust morphogenesis. Here, we review recent discoveries about the endogenous mechanisms of bioelectrical communication among non-neural cells that enables them to cooperate in vivo. We discuss important advances in bioelectronics, as well as computational and pharmacological tools that are enabling the taming of biophysical controls toward applications in regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA.
| | - John Selberg
- Electrical and Computer Engineering Department, University of California, Santa Cruz, CA 95064, USA
| | - Marco Rolandi
- Electrical and Computer Engineering Department, University of California, Santa Cruz, CA 95064, USA
| |
Collapse
|
23
|
Toward Decoding Bioelectric Events in Xenopus Embryogenesis: New Methodology for Tracking Interplay Between Calcium and Resting Potentials In Vivo. J Mol Biol 2019; 432:605-620. [PMID: 31711960 DOI: 10.1016/j.jmb.2019.10.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022]
Abstract
Although chemical signaling during embryogenesis is readily addressed by a plethora of available techniques, the developmental functions of ionic signaling are still poorly understood. It is increasingly realized that bioelectric events in nonneural cells are critical for pattern regulation, but their study has been hampered by difficulties in monitoring and manipulating them in vivo. Recent developments in visualizing electrical signaling dynamics in the field of neuroscience have facilitated functional experiments that reveal instructive developmental bioelectric signals. However, there is a pressing need for additional tools to explore time-dependent ionic signaling to understand complex endogenous dynamics. Here, we present methodological advances, including 4D imaging and data analysis, for improved tracking of calcium flux in the Xenopus laevis embryo, lowering the barrier for in vivo physiology work in this important model system. Using these techniques, we investigated the relationship between bioelectric ion channel activity and calcium, finding that cell hyperpolarization and depolarization both induce persistent static elevation of cytoplasmic calcium levels that fade over developmental time. These calcium changes correlate with increased cell mobility in early embryos and abnormal craniofacial morphology in later embryos. We thus highlight membrane potential modulation as a tractable tool for modulation of signaling cascades that rely on calcium as a transduction mechanism. The methods we describe facilitate the study of important novel aspects of developmental physiology, are extendable to numerous classes of existing and forthcoming fluorescent physiological reporters, and establish highly accessible, inexpensive protocols for their investigation.
Collapse
|
24
|
Djamgoz MBA, Fraser SP, Brackenbury WJ. In Vivo Evidence for Voltage-Gated Sodium Channel Expression in Carcinomas and Potentiation of Metastasis. Cancers (Basel) 2019; 11:E1675. [PMID: 31661908 PMCID: PMC6895836 DOI: 10.3390/cancers11111675] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/16/2022] Open
Abstract
A wide body of evidence suggests that voltage-gated sodium channels (VGSCs) are expressed de novo in several human carcinomas where channel activity promotes a variety of cellular behaviours integral to the metastatic cascade. These include directional motility (including galvanotaxis), pH balance, extracellular proteolysis, and invasion. Contrary to the substantial in vitro data, however, evidence for VGSC involvement in the cancer process in vivo is limited. Here, we critically assess, for the first time, the available in vivo evidence, hierarchically from mRNA level to emerging clinical aspects, including protein-level studies, electrolyte content, animal tests, and clinical imaging. The evidence strongly suggests that different VGSC subtypes (mainly Nav1.5 and Nav1.7) are expressed de novo in human carcinoma tissues and generally parallel the situation in vitro. Consistent with this, tissue electrolyte (sodium) levels, quantified by clinical imaging, are significantly higher in cancer vs. matched non-cancer tissues. These are early events in the acquisition of metastatic potential by the cancer cells. Taken together, the multi-faceted evidence suggests that the VGSC expression has clinical (diagnostic and therapeutic) potential as a prognostic marker, as well as an anti-metastatic target. The distinct advantages offered by the VGSC include especially (1) its embryonic nature, demonstrated most clearly for the predominant neonatal Nav1.5 expression in breast and colon cancer, and (2) the specifically druggable persistent current that VGSCs develop under hypoxic conditions, as in growing tumours, which promotes invasiveness and metastasis.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Scott P Fraser
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - William J Brackenbury
- Department of Biology and York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK.
| |
Collapse
|
25
|
Grossemy S, Chan PPY, Doran PM. Electrical stimulation of cell growth and neurogenesis using conductive and nonconductive microfibrous scaffolds. Integr Biol (Camb) 2019; 11:264-279. [DOI: 10.1093/intbio/zyz022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/01/2019] [Accepted: 06/12/2019] [Indexed: 11/13/2022]
Abstract
Abstract
The effect of exogenous electrical stimulation on cell viability, attachment, growth, and neurogenesis was examined using PC12 cells in microfibrous viscose-rayon scaffolds immersed in culture medium. The scaffolds were applied either in their nonconductive state or after coating the fibres with 200 nm of gold to give a scaffold sheet resistivity of (13 ± 1.3) Ω square−1. The cells were treated for 12 days using direct current electrical stimulation of 2 h per day. No cytotoxic effects were observed when up to 500 mV (8.3 mV mm−1) was applied to the scaffolds without gold, or when up to 100 mV (1.7 mV mm−1) was applied to the scaffolds with gold. Compared with unstimulated cells, whereas electrical stimulation significantly enhanced cell growth and attachment in the nonconductive scaffolds without gold, similar effects were not found for the conductive scaffolds with gold. Neural differentiation in the presence of nerve growth factor was improved by electrical stimulation in both scaffolds; however, neurite development and the expression of key differentiation markers were greater in the nonconductive scaffolds without gold than in the scaffolds with gold. Application of the same current to scaffolds with and without gold led to much higher levels of neurogenesis in the scaffolds without gold. This work demonstrates that substantial benefits in terms of cell growth and neural differentiation can be obtained using electric fields exerted across nonconductive microfibrous scaffolds, and that this approach to electrical stimulation can be more effective than when the stimulus is applied to cells on conductive scaffolds.
Collapse
Affiliation(s)
- Simon Grossemy
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, PO Box 218, Hawthorn, Melbourne, Australia
| | - Peggy P Y Chan
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, PO Box 218, Hawthorn, Melbourne, Australia
| | - Pauline M Doran
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, PO Box 218, Hawthorn, Melbourne, Australia
| |
Collapse
|
26
|
Cervera J, Pai VP, Levin M, Mafe S. From non-excitable single-cell to multicellular bioelectrical states supported by ion channels and gap junction proteins: Electrical potentials as distributed controllers. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 149:39-53. [PMID: 31255702 DOI: 10.1016/j.pbiomolbio.2019.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/26/2019] [Indexed: 12/18/2022]
Abstract
Endogenous bioelectric patterns within tissues are an important driver of morphogenesis and a tractable component of a number of disease states. Developing system-level understanding of the dynamics by which non-neural bioelectric circuits regulate complex downstream cascades is a key step towards both, an evolutionary understanding of ion channel genes, and novel strategies in regenerative medicine. An important capability gap is deriving rational modulation strategies targeting individual cells' bioelectric states to achieve global (tissue- or organ-level) outcomes. Here, we develop an ion channel-based model that describes multicellular states on the basis of spatio-temporal patterns of electrical potentials in aggregates of non-excitable cells. The model is of biological interest because modern techniques allow to associate bioelectrical signals with specific ion channel proteins in the cell membrane that are central to embryogenesis, regeneration, and tumorigenesis. As a complementary approach to the usual biochemical description, we have studied four biophysical questions: (i) how can single-cell bioelectrical states be established; (ii) how can a change in the cell potential caused by a transient perturbation of the cell state be maintained after the stimulus is gone (bioelectrical memory); (iii) how can a single-cell contribute to the control of multicellular ensembles based on the spatio-temporal pattern of electrical potentials; and (iv) how can oscillatory patterns arise from the single-cell bioelectrical dynamics. Experimentally, endogenous bioelectric gradients have emerged as instructive agents for morphogenetic processes. In this context, the simulations can guide new procedures that may allow a distributed control of the multicellular ensemble.
Collapse
Affiliation(s)
- Javier Cervera
- Dept. Termodinàmica, Universitat de València, E-46100, Burjassot, Spain.
| | - Vaibhav P Pai
- Dept. of Biology and Allen Discovery Center at Tufts University, Medford, MA, 02155-4243, USA
| | - Michael Levin
- Dept. of Biology and Allen Discovery Center at Tufts University, Medford, MA, 02155-4243, USA
| | - Salvador Mafe
- Dept. Termodinàmica, Universitat de València, E-46100, Burjassot, Spain
| |
Collapse
|
27
|
Yu S, Huang S, Ding Y, Wang W, Wang A, Lu Y. Transient receptor potential ion-channel subfamily V member 4: a potential target for cancer treatment. Cell Death Dis 2019; 10:497. [PMID: 31235786 PMCID: PMC6591233 DOI: 10.1038/s41419-019-1708-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/13/2019] [Accepted: 05/28/2019] [Indexed: 12/29/2022]
Abstract
The transient receptor potential ion-channel superfamily consists of nonselective cation channels located mostly on the plasma membranes of numerous animal cell types, which are closely related to sensory information transmission (e.g., vision, pain, and temperature perception), as well as regulation of intracellular Ca2+ balance and physiological activities of growth and development. Transient receptor potential ion channel subfamily V (TRPV) is one of the largest and most diverse subfamilies, including TRPV1-TRPV6 involved in the regulation of a variety of cellular functions. TRPV4 can be activated by various physical and chemical stimuli, such as heat, mechanical force, and phorbol ester derivatives participating in the maintenance of normal cellular functions. In recent years, the roles of TRPV4 in cell proliferation, differentiation, apoptosis, and migration have been extensively studied. Its abnormal expression has also been closely related to the onset and progression of multiple tumors, so TRPV4 may be a target for cancer diagnosis and treatment. In this review, we focused on the latest studies concerning the role of TRPV4 in tumorigenesis and the therapeutic potential. As evidenced by the effects on cancerogenesis, TRPV4 is a potential target for anticancer therapy.
Collapse
Affiliation(s)
- Suyun Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Shuai Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yushi Ding
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Wei Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
| |
Collapse
|
28
|
Ouyang D, Li R, Li Y, Zhu X. A 7-lncRNA signature predict prognosis of Uterine corpus endometrial carcinoma. J Cell Biochem 2019; 120:18465-18477. [PMID: 31168849 DOI: 10.1002/jcb.29164] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022]
Abstract
Current research indicate that long noncoding RNAs (lncRNAs) are associated with the progression of various cancers and can be used as prognostic biomarkers. This study aims to construct a prognostic lncRNA signature for the risk assessment of Uterine corpus endometrial carcinoma (UCEC). The RNA-Seq expression profile and corresponding clinical data of UCEC patients obtained from The Cancer Genome Atlas database. First, some prognosis-related lncRNAs were obtained by univariate Cox analysis. The minimum absolute contraction and selection operator (LASSO) regression and the Cox proportional hazard regression method were used to further identify the lncRNA prognostic model. Finally, seven lncRNAs (AC110491.1, AL451137.1, AC005381.1, AC103563.2, AC007422.2, AC108025.2, and MIR7-3HG) were identified as potential prognostic factors. According to the model constructed by the above analysis, the risk score of each UCEC patient was calculated, and the patients were classified into high and low-risk groups. The low-risk group had significant survival benefits. Moreover, we constructed a nomogram that incorporated independent prognostic factors (age, tumor stage, tumor grade, and risk score). The c-index value for evaluating the predictive nomogram model was 0.801. The area under the curve was 0.797 (3-year survival). The calibration curve also showed that there was a satisfactory agreement between the predicted and observed values in the probability of 1-, 3-, and 5-year overall survival. On the basis of the coexpression relationship, we established a coexpression network of lncRNA-messenger RNA (mRNA) of the 7-lncRNA. The Kyoto Encyclopedia of Genes and Genomes analysis of the coexpressing mRNAs showed that the main pathways related to the 7-lncRNA signature were neuroactive ligand-receptor interaction, serotonergic synapse, and gastric cancer pathway. Therefore, our study revealed that the 7-lncRNA could be used to predict the prognosis of UCEC and for postoperative treatment and follow-up.
Collapse
Affiliation(s)
- Dong Ouyang
- Department of Obstetrics and Gynecology, Akesu Hospital of Traditional Chinese Medicine, Akesu, China
| | - Ruyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaxian Li
- Department of Obstetrics and Gynecology, Akesu Hospital of Traditional Chinese Medicine, Akesu, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
29
|
D'Arcangelo D, Scatozza F, Giampietri C, Marchetti P, Facchiano F, Facchiano A. Ion Channel Expression in Human Melanoma Samples: In Silico Identification and Experimental Validation of Molecular Targets. Cancers (Basel) 2019; 11:cancers11040446. [PMID: 30934896 PMCID: PMC6520727 DOI: 10.3390/cancers11040446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/20/2019] [Accepted: 03/23/2019] [Indexed: 12/30/2022] Open
Abstract
Expression of 328 ion channel genes was investigated, by in silico analysis, in 170 human melanoma samples and controls. Ninety-one members of this gene-family (i.e., about 28%) show a significant (p < 0.05) differential expression in melanoma- vs. nevi-biopsies, taken from the GEO database. ROC (receiver operating characteristic) analysis selected 20 genes as potential markers showing the highest discrimination ability of melanoma vs. nevi (AUC > 0.90 and p < 0.0001). These 20 genes underwent a first in silico-validation round in an independent patients-dataset from GEO. A second-in silico-validation step was then carried out on a third human dataset in Oncomine. Finally, five genes were validated, showing extremely high sensitivity and specificity in melanoma detection (>90% in most cases). Such five genes (namely, SCNN1A, GJB3, KCNK7, GJB1, KCNN2) are novel potential melanoma markers or molecular targets, never previously related to melanoma. The “druggable genome” analysis was then carried out. Miconazole, an antifungal drug commonly used in clinics, is known to target KCNN2, the best candidate among the five identified genes. Miconazole was then tested in vitro in proliferation assays; it dose-dependently inhibited proliferation up to 90% and potently induced cell-death in A-375 and SKMEL-28 melanoma cells, while it showed no effect in control cells. Moreover, specific silencing of KCNN2 ion channel was achieved by siRNA transfection; under such condition miconazole strongly increases its anti-proliferative effect. In conclusion, the present study identified five ion channels that can potentially serve as sensitive and specific markers in human melanoma specimens and demonstrates that the antifungal drug miconazole, known to target one of the five identified ion channels, exerts strong and specific anti-melanoma effects in vitro.
Collapse
Affiliation(s)
| | | | - Claudia Giampietri
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy.
| | - Paolo Marchetti
- Medical Oncology, Sapienza, University of Rome, 00161 Rome, Italy.
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy.
| | - Antonio Facchiano
- Istituto Dermopatico dell'Immacolata (IDI-IRCCS), 00167 Rome, Italy.
| |
Collapse
|
30
|
Breuer EK, Fukushiro-Lopes D, Dalheim A, Burnette M, Zartman J, Kaja S, Wells C, Campo L, Curtis KJ, Romero-Moreno R, Littlepage LE, Niebur GL, Hoskins K, Nishimura MI, Gentile S. Potassium channel activity controls breast cancer metastasis by affecting β-catenin signaling. Cell Death Dis 2019; 10:180. [PMID: 30792401 PMCID: PMC6385342 DOI: 10.1038/s41419-019-1429-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Potassium ion channels are critical in the regulation of cell motility. The acquisition of cell motility is an essential parameter of cancer metastasis. However, the role of K+ channels in cancer metastasis has been poorly studied. High expression of the hG1 gene, which encodes for Kv11.1 channel associates with good prognosis in estrogen receptor-negative breast cancer (BC). We evaluated the efficacy of the Kv11.1 activator NS1643 in arresting metastasis in a triple negative breast cancer (TNBC) mouse model. NS1643 significantly reduces the metastatic spread of breast tumors in vivo by inhibiting cell motility, reprogramming epithelial–mesenchymal transition via attenuation of Wnt/β-catenin signaling and suppressing cancer cell stemness. Our findings provide important information regarding the clinical relevance of potassium ion channel expression in breast tumors and the mechanisms by which potassium channel activity can modulate tumor biology. Findings suggest that Kv11.1 activators may represent a novel therapeutic approach for the treatment of metastatic estrogen receptor-negative BC. Ion channels are critical factor for cell motility but little is known about their role in metastasis. Stimulation of the Kv11.1 channel suppress the metastatic phenotype in TNBC. This work could represent a paradigm-shifting approach to reducing mortality by targeting a pathway that is central to the development of metastases.
Collapse
Affiliation(s)
- Eun-Kyoung Breuer
- Department of Radiation Oncology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Daniela Fukushiro-Lopes
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Annika Dalheim
- Department of Surgery, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Miranda Burnette
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Simon Kaja
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Claire Wells
- Division of Cancer Studies, King's College London, Rm. 2.34 A New Hunts House, Guy's Campus, London, SE1 1 UL, UK
| | - Loredana Campo
- Department of Radiation Oncology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Kimberly J Curtis
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Ricardo Romero-Moreno
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Laurie E Littlepage
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Glen L Niebur
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA.,Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Kent Hoskins
- Division of Hematology Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Michael I Nishimura
- Department of Surgery, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Saverio Gentile
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA. .,Division of Hematology Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
31
|
Zhang G, Wang X, Xue Q. Combined targeted ion channel therapy: Can it be an alternative choice for esophageal cancer patients? Med Hypotheses 2018; 117:59-62. [DOI: 10.1016/j.mehy.2018.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 04/23/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023]
|