1
|
Djamgoz MBA. Stemness of Cancer: A Study of Triple-negative Breast Cancer From a Neuroscience Perspective. Stem Cell Rev Rep 2025; 21:337-350. [PMID: 39531198 PMCID: PMC11872763 DOI: 10.1007/s12015-024-10809-0] [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] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Stemness, giving cancer cells massive plasticity enabling them to survive in dynamic (e.g. hypoxic) environments and become resistant to treatment, especially chemotherapy, is an important property of aggressive tumours. Here, we review some essentials of cancer stemness focusing on triple-negative breast cancer (TNBC), the most aggressive form of all breast cancers. TNBC cells express a range of genes and mechanisms associated with stemness, including the fundamental four "Yamanaka factors". Most of the evidence concerns the transcription factor / oncogene c-Myc and an interesting case is the expression of the neonatal splice variant of voltage-gated sodium channel subtype Nav1.5. On the whole, measures that reduce the stemness make cancer cells less aggressive, reducing their invasive/metastatic potential and increasing/restoring their chemosensitivity. Such measures include gene silencing techniques, epigenetic therapies as well as novel approaches like optogenetics aiming to modulate the plasma membrane voltage. Indeed, simply hyperpolarizing their membrane potential can make stem cells differentiate. Finally, we give an overview of the clinical aspects and exploitation of cancer/TNBC stemness, including diagnostics and therapeutics. In particular, personalised mRNA-based therapies and mechanistically meaningful combinations are promising and the emerging discipline of 'cancer neuroscience' is providing novel insights to both fundamental issues and clinical applications.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
2
|
Shi Q, Yang Z, Yang H, Xu L, Xia J, Gu J, Chen M, Wang Y, Zhao X, Liao Z, Mou Y, Gu X, Xie T, Sui X. Targeting ion channels: innovative approaches to combat cancer drug resistance. Theranostics 2025; 15:521-545. [PMID: 39744692 PMCID: PMC11671388 DOI: 10.7150/thno.103384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
Ion channels, as functional molecules that regulate the flow of ions across cell membranes, have emerged as a promising target in cancer therapy due to their pivotal roles in cell proliferation, metastasis, apoptosis, drug resistance, and so on. Recently, increasing evidence suggests that dysregulation of ion channels is a common characteristic of cancer cells, contributing to their survival and the resistance to conventional therapies. For example, the aberrant expression of sodium (Na+) and potassium ion (K+) channels is significantly correlated with the sensitivity of chemotherapy drugs. The endogenous calcium (Ca2+) channels contribute to the acquired resistance of osimertinib in epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer cell lines. Ferrous ions (Fe2+) enhance the sensitivity of breast cancer cells to doxorubicin treatment. Preclinical models have also demonstrated the effect of specific ion channel blockers or modulators on anticancer drug resistance. This review describes the current understanding about the interaction between ion channels and the therapeutic efficacy of anticancer drugs. Then, the therapeutic potential of ion channel blockers or modulators in enhancing the sensitivity or overcoming the resistance of cancer cells to anticancer therapies is discussed. Targeting ion channels will hopefully offer a novel and promising strategy for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Qian Shi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zijing Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lihui Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jing Xia
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jie Gu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengting Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yan Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaohong Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zehua Liao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yiping Mou
- General Surgery, Cancer Center, Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical University, Hangzhou, Zhejiang, China
| | - Xidong Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
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
|
4
|
Giammello F, Biella C, Priori EC, Filippo MADS, Leone R, D'Ambrosio F, Paterno' M, Cassioli G, Minetti A, Macchi F, Spalletti C, Morella I, Ruberti C, Tremonti B, Barbieri F, Lombardi G, Brambilla R, Florio T, Galli R, Rossi P, Brandalise F. Modulating voltage-gated sodium channels to enhance differentiation and sensitize glioblastoma cells to chemotherapy. Cell Commun Signal 2024; 22:434. [PMID: 39251990 PMCID: PMC11382371 DOI: 10.1186/s12964-024-01819-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) stands as the most prevalent and aggressive form of adult gliomas. Despite the implementation of intensive therapeutic approaches involving surgery, radiation, and chemotherapy, Glioblastoma Stem Cells contribute to tumor recurrence and poor prognosis. The induction of Glioblastoma Stem Cells differentiation by manipulating the transcriptional machinery has emerged as a promising strategy for GBM treatment. Here, we explored an innovative approach by investigating the role of the depolarized resting membrane potential (RMP) observed in patient-derived GBM sphereforming cell (GSCs), which allows them to maintain a stemness profile when they reside in the G0 phase of the cell cycle. METHODS We conducted molecular biology and electrophysiological experiments, both in vitro and in vivo, to examine the functional expression of the voltage-gated sodium channel (Nav) in GSCs, particularly focusing on its cell cycle-dependent functional expression. Nav activity was pharmacologically manipulated, and its effects on GSCs behavior were assessed by live imaging cell cycle analysis, self-renewal assays, and chemosensitivity assays. Mechanistic insights into the role of Nav in regulating GBM stemness were investigated through pathway analysis in vitro and through tumor proliferation assay in vivo. RESULTS We demonstrated that Nav is functionally expressed by GSCs mainly during the G0 phase of the cell cycle, suggesting its pivotal role in modulating the RMP. The pharmacological blockade of Nav made GBM cells more susceptible to temozolomide (TMZ), a standard drug for this type of tumor, by inducing cell cycle re-entry from G0 phase to G1/S transition. Additionally, inhibition of Nav substantially influenced the self-renewal and multipotency features of GSCs, concomitantly enhancing their degree of differentiation. Finally, our data suggested that Nav positively regulates GBM stemness by depolarizing the RMP and suppressing the ERK signaling pathway. Of note, in vivo proliferation assessment confirmed the increased susceptibility to TMZ following pharmacological blockade of Nav. CONCLUSIONS This insight positions Nav as a promising prognostic biomarker and therapeutic target for GBM patients, particularly in conjunction with temozolomide treatment.
Collapse
Affiliation(s)
- Francesca Giammello
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
- PhD Program in Genetics, Molecular and Cellular Biology, University of Pavia, Pavia, Italy
| | - Chiara Biella
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Roberta Leone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Martina Paterno'
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Giulia Cassioli
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Antea Minetti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Francesca Macchi
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Cristina Spalletti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Ilaria Morella
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Cristina Ruberti
- Advanced Technology Platform, Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Beatrice Tremonti
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Federica Barbieri
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Giuseppe Lombardi
- Department of Oncology 1, Oncology, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, Padua, 35128, Italy
| | - Riccardo Brambilla
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Tullio Florio
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Rossella Galli
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | |
Collapse
|
5
|
Bianconi S, Leppik L, Oppermann E, Marzi I, Henrich D. Direct Current Electrical Stimulation Shifts THP-1-Derived Macrophage Polarization towards Pro-Regenerative M2 Phenotype. Int J Mol Sci 2024; 25:7272. [PMID: 39000377 PMCID: PMC11242703 DOI: 10.3390/ijms25137272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
A macrophage shift from the M1 to the M2 phenotype is relevant for promoting tissue repair and regeneration. In a previous in vivo study, we found that direct current (DC) electrical stimulation (EStim) increased the proportion of M2 macrophages in healing tissues and directed the balance of the injury response away from healing/scarring towards regeneration. These observations led us to hypothesize that DC EStim regulates macrophage polarization towards an M2 phenotype. THP-1-derived M0, M1 (IFN-γ and LPS), and M2 (IL-4 and IL-13) macrophages were exposed (or not: control group) to 100 mV/mm of DC EStim, 1 h/day for three days. Macrophage polarization was assessed through gene and surface marker expressions and cytokine secretion profiles. Following DC EStim treatment, M0 cells exhibited an upregulation of M2 marker genes IL10, CD163, and PPARG. In M1 cells, DC EStim upregulated the gene expressions of M2 markers IL10, TGM2, and CD206 and downregulated M1 marker gene CD86. EStim treatment also reduced the surface expression of CD86 and secretion of pro-inflammatory cytokines IL-1β and IL-6. Our results suggest that DC EStim differentially exerts pro-M2 effects depending on the macrophage phenotype: it upregulates typical M2 genes in M0 and M1 cells while inhibiting M1 marker CD86 at the nuclear and protein levels and the secretion of pro-inflammatory interleukins in M1 cells. Conversely, M2 cells appear to be less responsive to the EStim treatment employed in this study.
Collapse
Affiliation(s)
- Santiago Bianconi
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Liudmila Leppik
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Elsie Oppermann
- Department of General, Visceral, Transplant and Thoracic Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Ingo Marzi
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| |
Collapse
|
6
|
Pio-Lopez L, Levin M. Aging as a loss of morphostatic information: A developmental bioelectricity perspective. Ageing Res Rev 2024; 97:102310. [PMID: 38636560 DOI: 10.1016/j.arr.2024.102310] [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: 11/05/2023] [Revised: 02/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Maintaining order at the tissue level is crucial throughout the lifespan, as failure can lead to cancer and an accumulation of molecular and cellular disorders. Perhaps, the most consistent and pervasive result of these failures is aging, which is characterized by the progressive loss of function and decline in the ability to maintain anatomical homeostasis and reproduce. This leads to organ malfunction, diseases, and ultimately death. The traditional understanding of aging is that it is caused by the accumulation of molecular and cellular damage. In this article, we propose a complementary view of aging from the perspective of endogenous bioelectricity which has not yet been integrated into aging research. We propose a view of aging as a morphostasis defect, a loss of biophysical prepattern information, encoding anatomical setpoints used for dynamic tissue and organ homeostasis. We hypothesize that this is specifically driven by abrogation of the endogenous bioelectric signaling that normally harnesses individual cell behaviors toward the creation and upkeep of complex multicellular structures in vivo. Herein, we first describe bioelectricity as the physiological software of life, and then identify and discuss the links between bioelectricity and life extension strategies and age-related diseases. We develop a bridge between aging and regeneration via bioelectric signaling that suggests a research program for healthful longevity via morphoceuticals. Finally, we discuss the broader implications of the homologies between development, aging, cancer and regeneration and how morphoceuticals can be developed for aging.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Urazaev AK, Wang L, Bai Y, Adissu HA, Lelièvre SA. The epithelial polarity axis controls the resting membrane potential and Cl- co-transport in breast glandular structures. J Cell Sci 2024; 137:jcs260924. [PMID: 37818620 PMCID: PMC10651101 DOI: 10.1242/jcs.260924] [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: 12/30/2022] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
The membrane potential (MP) controls cell homeostasis by directing molecule transport and gene expression. How the MP is set upon epithelial differentiation is unknown. Given that tissue architecture also controls homeostasis, we investigated the relationship between basoapical polarity and resting MP in three-dimensional culture of the HMT-3522 breast cancer progression. A microelectrode technique to measure MP and input resistance reveals that the MP is raised by gap junction intercellular communication (GJIC), which directs tight-junction mediated apical polarity, and is decreased by the Na+/K+/2Cl- (NKCC, encoded by SLC12A1 and SLC12A2) co-transporter, active in multicellular structures displaying basal polarity. In the tumor counterpart, the MP is reduced. Cancer cells display diminished GJIC and do not respond to furosemide, implying loss of NKCC activity. Induced differentiation of cancer cells into basally polarized multicellular structures restores widespread GJIC and NKCC responses, but these structures display the lowest MP. The absence of apical polarity, necessary for cancer onset, in the non-neoplastic epithelium is also associated with the lowest MP under active Cl- transport. We propose that the loss of apical polarity in the breast epithelium destabilizes cellular homeostasis in part by lowering the MP.
Collapse
Affiliation(s)
- Albert K. Urazaev
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- School of Liberal Arts, Sciences and Education, Ivy Tech Community College, Lafayette, IN 47905, USA
| | - Lei Wang
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Yunfeng Bai
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, West Lafayette, IN 47907, USA
| | - Hibret A. Adissu
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Sophie A. Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, West Lafayette, IN 47907, USA
- Relation Gene-Environment-REGEN Unit, Institut de Cancérologie de l'Ouest (ICO), Angers 49055, France
| |
Collapse
|
8
|
Zeng X, Yang Y. Molecular Mechanisms Underlying Vascular Remodeling in Hypertension. Rev Cardiovasc Med 2024; 25:72. [PMID: 39077331 PMCID: PMC11263180 DOI: 10.31083/j.rcm2502072] [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: 07/05/2023] [Revised: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 07/31/2024] Open
Abstract
Hypertension, a common cardiovascular disease, is primarily characterized by vascular remodeling. Recent extensive research has led to significant progress in understanding its mechanisms. Traditionally, vascular remodeling has been described as a unidirectional process in which blood vessels undergo adaptive remodeling or maladaptive remodeling. Adaptive remodeling involves an increase in vessel diameter in response to increased blood flow, while maladaptive remodeling refers to the narrowing or thickening of blood vessels in response to pathological conditions. However, recent research has revealed that vascular remodeling is much more complex. It is now understood that vascular remodeling is a dynamic interplay between various cellular and molecular events. This interplay process involves different cell types, including endothelial cells, smooth muscle cells, and immune cells, as well as their interactions with the extracellular matrix. Through these interactions, blood vessels undergo intricate and dynamic changes in structure and function in response to various stimuli. Moreover, vascular remodeling involves various factors and mechanisms such as the renin-angiotensin-aldosterone system (RAS), oxidative stress, inflammation, the extracellular matrix (ECM), sympathetic nervous system (SNS) and mechanical stress that impact the arterial wall. These factors may lead to vascular and circulatory system diseases and are primary causes of long-term increases in systemic vascular resistance in hypertensive patients. Additionally, the presence of stem cells in adventitia, media, and intima of blood vessels plays a crucial role in vascular remodeling and disease development. In the future, research will focus on examining the underlying mechanisms contributing to hypertensive vascular remodeling to develop potential solutions for hypertension treatment. This review provides us with a fresh perspective on hypertension and vascular remodeling, undoubtedly sparking further research efforts aimed at uncovering more potent treatments and enhanced preventive and control measures for this disease.
Collapse
Affiliation(s)
- Xinyi Zeng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
9
|
Masuelli S, Real S, McMillen P, Oudin M, Levin M, Roqué M. The Yin and Yang of Breast Cancer: Ion Channels as Determinants of Left-Right Functional Differences. Int J Mol Sci 2023; 24:11121. [PMID: 37446299 PMCID: PMC10342022 DOI: 10.3390/ijms241311121] [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: 05/11/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer is a complex and heterogeneous disease that displays diverse molecular subtypes and clinical outcomes. Although it is known that the location of tumors can affect their biological behavior, the underlying mechanisms are not fully understood. In our previous study, we found a differential methylation profile and membrane potential between left (L)- and right (R)-sided breast tumors. In this current study, we aimed to identify the ion channels responsible for this phenomenon and determine any associated phenotypic features. To achieve this, experiments were conducted in mammary tumors in mice, human patient samples, and with data from public datasets. The results revealed that L-sided tumors have a more depolarized state than R-sided. We identified a 6-ion channel-gene signature (CACNA1C, CACNA2D2, CACNB2, KCNJ11, SCN3A, and SCN3B) associated with the side: L-tumors exhibit lower expression levels than R-tumors. Additionally, in silico analyses show that the signature correlates inversely with DNA methylation writers and with key biological processes involved in cancer progression, such as proliferation and stemness. The signature also correlates inversely with patient survival rates. In an in vivo mouse model, we confirmed that KI67 and CD44 markers were increased in L-sided tumors and a similar tendency for KI67 was found in patient L-tumors. Overall, this study provides new insights into the potential impact of anatomical location on breast cancer biology and highlights the need for further investigation into possible differential treatment options.
Collapse
Affiliation(s)
- Sofía Masuelli
- Institute of Histology and Embryology, National Council of Scientific and Technological Research (CONICET), Parque General San Martin, Mendoza 5500, Argentina; (S.M.)
- Faculty of Medical Science, National University of Cuyo, Parque General San Martin, Mendoza 5500, Argentina
| | - Sebastián Real
- Institute of Histology and Embryology, National Council of Scientific and Technological Research (CONICET), Parque General San Martin, Mendoza 5500, Argentina; (S.M.)
- Faculty of Medical Science, National University of Cuyo, Parque General San Martin, Mendoza 5500, Argentina
| | - Patrick McMillen
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Madeleine Oudin
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - María Roqué
- Institute of Histology and Embryology, National Council of Scientific and Technological Research (CONICET), Parque General San Martin, Mendoza 5500, Argentina; (S.M.)
- Faculty of Exact and Natural Sciences, National University of Cuyo, Parque General San Martin, Mendoza 5500, Argentina
| |
Collapse
|
10
|
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
|
11
|
Liao K, Cui Z, Wang Z, Peng Y, Tang S, Chen J. Hyperosmolar Potassium Inhibits Corneal Myofibroblast Transformation and Prevent Corneal Scar. Curr Eye Res 2023; 48:238-250. [PMID: 36149345 DOI: 10.1080/02713683.2022.2129072] [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] [Indexed: 11/03/2022]
Abstract
PURPOSE Corneal myofibroblasts play a crucial role in the process of corneal scarring. Potassium has been documented to reduce skin scar tissue formation. Herein, we investigated the ability of potassium to prevent corneal fibrosis in cell culture and in vivo. METHODS Corneal fibroblasts (CFs) were isolated from the corneal limbus and treated with TGF-β1 to transform into corneal myofibroblasts. Corneal myofibroblast markers were detected by quantitative real-time PCR, Western blot, and immunofluorescence. The contractive functions of corneal myofibroblast were evaluated by the scratch assay and the collagen gel contraction assay. RNA sequencing in corneal fibroblasts was performed to explore the mechanisms underlying hyperosmolar potassium treatment. GO and KEGG analysis were performed to explore the underlying mechanism by hyperosmolar potassium treatment. The ATP detection assay assessed the level of cell metabolism. KCl eye drops four times per day were administered to mice models of corneal injury to evaluate the ability to prevent corneal scar formation. Corneal opacity area was evaluated by Image J software. RESULTS Treatment with hyperosmolar potassium could suppress corneal myofibroblast transformation and collagen I synthesis induced by TGF-β1 in cell culture. Hyperosmolar potassium could inhibit wound healing and gel contraction in CFs. RNA sequencing results suggested that genes involved in the metabolic pathway were downregulated after KCl treatment. ATP levels were significantly decreased in the KCl group compared with the control group. Hyperosmolar potassium could prevent corneal myofibroblast transformation after corneal injury and corneal scar formation in mice. CONCLUSION Potassium can suppress corneal myofibroblast transformation and collagen I protein synthesis. Moreover, given that KCl eye drops can prevent corneal scar formation, it has been suggested to have huge prospects as a novel treatment approach during clinical practice.
Collapse
Affiliation(s)
- Kai Liao
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Zekai Cui
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Zhijie Wang
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Yu Peng
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiansu Chen
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Sempou E, Kostiuk V, Zhu J, Cecilia Guerra M, Tyan L, Hwang W, Camacho-Aguilar E, Caplan MJ, Zenisek D, Warmflash A, Owens NDL, Khokha MK. Membrane potential drives the exit from pluripotency and cell fate commitment via calcium and mTOR. Nat Commun 2022; 13:6681. [PMID: 36335122 PMCID: PMC9637099 DOI: 10.1038/s41467-022-34363-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/24/2022] [Indexed: 11/08/2022] Open
Abstract
Transitioning from pluripotency to differentiated cell fates is fundamental to both embryonic development and adult tissue homeostasis. Improving our understanding of this transition would facilitate our ability to manipulate pluripotent cells into tissues for therapeutic use. Here, we show that membrane voltage (Vm) regulates the exit from pluripotency and the onset of germ layer differentiation in the embryo, a process that affects both gastrulation and left-right patterning. By examining candidate genes of congenital heart disease and heterotaxy, we identify KCNH6, a member of the ether-a-go-go class of potassium channels that hyperpolarizes the Vm and thus limits the activation of voltage gated calcium channels, lowering intracellular calcium. In pluripotent embryonic cells, depletion of kcnh6 leads to membrane depolarization, elevation of intracellular calcium levels, and the maintenance of a pluripotent state at the expense of differentiation into ectodermal and myogenic lineages. Using high-resolution temporal transcriptome analysis, we identify the gene regulatory networks downstream of membrane depolarization and calcium signaling and discover that inhibition of the mTOR pathway transitions the pluripotent cell to a differentiated fate. By manipulating Vm using a suite of tools, we establish a bioelectric pathway that regulates pluripotency in vertebrates, including human embryonic stem cells.
Collapse
Affiliation(s)
- Emily Sempou
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Valentyna Kostiuk
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Jie Zhu
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - M Cecilia Guerra
- Departments of Biosciences and Bioengineering Rice University, 345 Anderson Biological Labs, Houston, TX, 77005, USA
| | - Leonid Tyan
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Woong Hwang
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Elena Camacho-Aguilar
- Departments of Biosciences and Bioengineering Rice University, 345 Anderson Biological Labs, Houston, TX, 77005, USA
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - David Zenisek
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Aryeh Warmflash
- Departments of Biosciences and Bioengineering Rice University, 345 Anderson Biological Labs, Houston, TX, 77005, USA
| | - Nick D L Owens
- Department of Clinical and Biomedical Sciences, University of Exeter, Barrack Road, Exeter, EX2 5DW, UK
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
| |
Collapse
|
13
|
Bioelectronic medicines: Therapeutic potential and advancements in next-generation cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188808. [DOI: 10.1016/j.bbcan.2022.188808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
|
14
|
Carvalho J. A computational model of organism development and carcinogenesis resulting from cells' bioelectric properties and communication. Sci Rep 2022; 12:9206. [PMID: 35654933 PMCID: PMC9163332 DOI: 10.1038/s41598-022-13281-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/23/2022] [Indexed: 11/15/2022] Open
Abstract
A sound theory of biological organization is clearly missing for a better interpretation of observational results and faster progress in understanding life complexity. The availability of such a theory represents a fundamental progress in explaining both normal and pathological organism development. The present work introduces a computational implementation of some principles of a theory of organism development, namely that the default state of cells is proliferation and motility, and includes the principle of variation and organization by closure of constraints. In the present model, the bioelectric context of cells and tissue is the field responsible for organization, as it regulates cell proliferation and the level of communication driving the system's evolution. Starting from a depolarized (proliferative) cell, the organism grows to a certain size, limited by the increasingly polarized state after successive proliferation events. The system reaches homeostasis, with a depolarized core (proliferative cells) surrounded by a rim of polarized cells (non-proliferative in this condition). This state is resilient to cell death (random or due to injure) and to limited depolarization (potentially carcinogenic) events. Carcinogenesis is introduced through a localized event (a spot of depolarized cells) or by random depolarization of cells in the tissue, which returns cells to their initial proliferative state. The normalization of the bioelectric condition can reverse this out-of-equilibrium state to a new homeostatic one. This simplified model of embryogenesis, tissue organization and carcinogenesis, based on non-excitable cells' bioelectric properties, can be made more realistic with the introduction of other components, like biochemical fields and mechanical interactions, which are fundamental for a more faithful representation of reality. However, even a simple model can give insight for new approaches in complex systems and suggest new experimental tests, focused in its predictions and interpreted under a new paradigm.
Collapse
Affiliation(s)
- Joao Carvalho
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
15
|
Davidian D, Levin M. Inducing Vertebrate Limb Regeneration: A Review of Past Advances and Future Outlook. Cold Spring Harb Perspect Biol 2022; 14:a040782. [PMID: 34400551 PMCID: PMC9121900 DOI: 10.1101/cshperspect.a040782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Limb loss due to traumatic injury or amputation is a major biomedical burden. Many vertebrates exhibit the ability to form and pattern normal limbs during embryogenesis from amorphous clusters of precursor cells, hinting that this process could perhaps be activated later in life to rebuild missing or damaged limbs. Indeed, some animals, such as salamanders, are proficient regenerators of limbs throughout their life span. Thus, research over the last century has sought to stimulate regeneration in species that do not normally regenerate their appendages. Importantly, these efforts are not only a vital aspect of regenerative medicine, but also have fundamental implications for understanding evolution and the cellular control of growth and form throughout the body. Here we review major recent advances in augmenting limb regeneration, summarizing the degree of success that has been achieved to date in frog and mammalian models using genetic, biochemical, and bioelectrical interventions. While the degree of whole limb repair in rodent models has been modest to date, a number of new technologies and approaches comprise an exciting near-term road map for basic and clinical progress in regeneration.
Collapse
Affiliation(s)
- Devon Davidian
- Allen Discovery Center at Tufts University, Medford, Massachusetts 02155, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, Massachusetts 02155, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA
| |
Collapse
|
16
|
Potier-Cartereau M, Gautier M, Ravalet N, Ducrocq E, Hamard S, LeGuennec JY, Vandier C, Herault O. The Sodium-Calcium Exchanger Controls the Membrane Potential of AFT024: A Mesenchymal Stem Cell Hematopoietic Niche Forming Line. Bioelectricity 2022; 4:103-107. [PMID: 39350778 PMCID: PMC11441364 DOI: 10.1089/bioe.2022.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to characterize the functional expression of sodium-calcium exchangers on AFT024 cell line, a murine model of mesenchymal stem/stromal cells (MSCs) supporting human primitive hematopoiesis. All current-clamp and voltage-clamp experiments were performed using the perforated patch whole-cell recording technique with amphotericin B. The membrane potential of -14 mV shifted to -35 mV when lowering the external sodium concentration to 0.33 mM and an increase of cytosolic calcium concentration was observed. KB-R7943, a selective blocker of cardiac sodium-calcium exchangers, also named NCX1, induced a hyperpolarization at physiological sodium concentration while it blocked the hyperpolarization observed at low sodium concentration. This demonstrates for the first time the presence of the sodium-calcium exchangers in AFT024 cells and provides initial evidence that the membrane potential of these stromal cells is maintained depolarized by this exchanger. Lowering external sodium concentration and KB-R7943 had no effect on the membrane potential of 2018 cells, a nonhematopoietic-supportive cell line. Since NCX1 is differentially expressed in AFT024 cells as compared with nonhematopoietic supportive cells with more restricted differentiation potential, this study suggests a potential role of this sodium-calcium exchanger, in the differentiation process or hematopoietic support of MSCs.
Collapse
Affiliation(s)
| | - Mathieu Gautier
- LPCM UR 4667, University of Picardie Jules Verne, Amiens, France
| | - Noemie Ravalet
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
| | - Elfi Ducrocq
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
| | - Sophie Hamard
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
| | - Jean-Yves LeGuennec
- UMR 01046–UMR 9214, University of Montpellier, INSERM, CNRS, Montpellier, France
| | | | - Olivier Herault
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
- Department of Biological Hematology, Tours University Hospital, Tours, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
17
|
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
|
18
|
Hornsby E, King HW, Peiris M, Buccafusca R, Lee WYJ, Wing ES, Blackshaw LA, Lindsay JO, Stagg AJ. The cation channel TRPM8 influences the differentiation and function of human monocytes. J Leukoc Biol 2022; 112:365-381. [PMID: 35233801 PMCID: PMC9543907 DOI: 10.1002/jlb.1hi0421-181r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
Monocytes are mononuclear phagocytes that can differentiate to a variety of cell fates under the influence of their microenvironment and hardwired commitment. We found that inhibition of TRPM8 in human blood CD14+ monocytes during a critical 3‐h window at the beginning of their differentiation into macrophages led to enhanced survival and LPS‐driven TNFα production after 24 h. TRPM8 antagonism also promoted LPS‐driven TNFα production in CD14+ monocytes derived from the intestinal mucosa. Macrophages that had been derived for 6 days under blockade of TRPM8 had impaired phagocytic capacity and were transcriptionally distinct. Most of the affected genes were altered in a way that opposed normal monocyte to macrophage differentiation indicating that TRPM8 activity promotes aspects of this differentiation programme. Thus, we reveal a novel role for TRPM8 in regulating human CD14+ monocyte fate and function.
Collapse
Affiliation(s)
- Eve Hornsby
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Hamish W King
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Madusha Peiris
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Roberto Buccafusca
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, UK
| | - Wing-Yiu Jason Lee
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Elinor S Wing
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - L Ashley Blackshaw
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - James O Lindsay
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Department of Gastroenterology, Barts Health NHS Trust, The Royal London Hospital, Whitechapel, London, UK
| | - Andrew J Stagg
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
19
|
Zhang M, Che C, Cheng J, Li P, Yang Y. Ion channels in stem cells and their roles in stem cell biology and vascular diseases. J Mol Cell Cardiol 2022; 166:63-73. [PMID: 35143836 DOI: 10.1016/j.yjmcc.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
Abstract
Stem cell therapy may be a promising option for the treatment of vascular diseases. In recent years, significant progress has been made in stem cell research, especially in the mechanism of stem cell activation, homing and differentiation in vascular repair and reconstruction. Current research on stem cells focuses on protein expression and transcriptional networks. Ion channels are considered to be the basis for the generation of bioelectrical signals, which control the proliferation, differentiation and migration of various cell types. Although heterogeneity of multiple ion channels has been found in different types of stem cells, it is unclear whether the heterogeneous expression of ion channels is related to different cell subpopulations and/or different stages of the cell cycle. There is still a long way to go in clinical treatment by using the regulation of stem cell ion channels. In this review, we reviewed the main ion channels found on stem cells, their expression and function in stem cell proliferation, differentiation and migration, and the research status of stem cells' involvement in vascular diseases.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Chang Che
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| |
Collapse
|
20
|
Chang CY, Park JH, Ouh IO, Gu NY, Jeong SY, Lee SA, Lee YH, Hyun BH, Kim KS, Lee J. Novel method to repair articular cartilage by direct reprograming of prechondrogenic mesenchymal stem cells. Eur J Pharmacol 2021; 911:174416. [PMID: 34606836 DOI: 10.1016/j.ejphar.2021.174416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022]
Abstract
Age-related cartilage loss is worsened by the limited regenerative capacity of chondrocytes. The role of cell-based therapies using mesenchymal stem cells is gaining interest. Adipose tissue-derived mesenchymal stem cells (ADSCs) are an attractive source to generate the optimal number of chondrocytes required to repair a cartilage defect and regenerate hyaline articular cartilage. Here, we report an outstanding technique to prepare chondrocytes for cartilage repair using canine ADSCs. We hypothesized that external electrical fields promote prechondrogenic condensation without requiring genetic modifications or exogenous factors. We analyzed the effect of electrical stimulation (ES) on the differentiation of ADSC micromass into chondrocytes. Highly compact structures were formed within 3 days of ES of canine ADSC micromass. The expression of type I collagen gene was abolished in these cells compared with that in control micromass cultures and monolayer cultures. We further found that ES enhanced the production of proteoglycan, a highly produced extracellular matrix component in chondrocytes. Additionally, single-cell RNA sequencing analysis showed that canine ADSC micromass undergoing ES developed a prechondrogenic cell aggregation, suggesting their metabolic conversion, biogenesis, and calcium ion change. Collectively, our findings demonstrate the capacity of ES to drive the chondrogenesis of ADSCs in the absence of exogenous factors and confirm its commercial potential as a budget-friendly therapy for the repair of cartilage defects.
Collapse
Affiliation(s)
- Chi Young Chang
- Hanyang Digitech, 332-7, Samsung 1-ro, Hwaseong, Gyeonggi-do, 18380, Republic of Korea; Youth Bio Global, 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea
| | - Ju Hyun Park
- Hanyang Digitech, 332-7, Samsung 1-ro, Hwaseong, Gyeonggi-do, 18380, Republic of Korea; Youth Bio Global, 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea
| | - In-Ohk Ouh
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - So Yeon Jeong
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Se-A Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Yoon-Hee Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Bang-Hun Hyun
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Ki Suk Kim
- Hanyang Digitech, 332-7, Samsung 1-ro, Hwaseong, Gyeonggi-do, 18380, Republic of Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea; Division of Regenerative Medicine Safety Control, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, 187 Osongsaengmyeong 2-ro, Cheongju, Chungcheongbuk-do, 28159, Republic of Korea.
| |
Collapse
|
21
|
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
|
22
|
Cui X, Li X, He Y, Yu J, Dong N, Zhao RC. Slight up-regulation of Kir2.1 channel promotes endothelial progenitor cells to transdifferentiate into a pericyte phenotype by Akt/mTOR/Snail pathway. J Cell Mol Med 2021; 25:10088-10100. [PMID: 34592781 PMCID: PMC8572793 DOI: 10.1111/jcmm.16944] [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: 08/19/2020] [Revised: 08/22/2021] [Accepted: 09/19/2021] [Indexed: 12/27/2022] Open
Abstract
It was shown that endothelial progenitor cells (EPCs) have bidirectional differentiation potential and thus perform different biological functions. The purpose of this study was to investigate the effects of slight up‐regulation of the Kir2.1 channel on EPC transdifferentiation and the potential mechanism on cell function and transformed cell type. First, we found that the slight up‐regulation of Kir2.1 expression promoted the expression of the stem cell stemness factors ZFX and NS and inhibited the expression of senescence‐associated β‐galactosidase. Further studies showed the slightly increased expression of Kir2.1 could also improve the expression of pericyte molecular markers NG2, PDGFRβ and Desmin. Moreover, adenovirus‐mediated Kir2.1 overexpression had an enhanced contractile response to norepinephrine of EPCs. These results suggest that the up‐regulated expression of the Kir2.1 channel promotes EPC transdifferentiation into a pericyte phenotype. Furthermore, the mechanism of EPC transdifferentiation to mesenchymal cells (pericytes) was found to be closely related to the channel functional activity of Kir2.1 and revealed that this channel could promote EPC EndoMT by activating the Akt/mTOR/Snail signalling pathway. Overall, this study suggested that in the early stage of inflammatory response, regulating the Kir2.1 channel expression affects the biological function of EPCs, thereby determining the maturation and stability of neovascularization.
Collapse
Affiliation(s)
- Xiaodong Cui
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China.,School of Basic Medicine Sciences, Weifang Medical University, Weifang, China
| | - Xiaoxia Li
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Yanting He
- School of Basic Medicine Sciences, Weifang Medical University, Weifang, China
| | - Jie Yu
- School of Basic Medicine Sciences, Weifang Medical University, Weifang, China
| | - Naijun Dong
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Robert Chunhua Zhao
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Grodstein J, Levin M. Stability and robustness properties of bioelectric networks: A computational approach. BIOPHYSICS REVIEWS 2021; 2:031305. [PMID: 38505634 PMCID: PMC10903393 DOI: 10.1063/5.0062442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/07/2021] [Indexed: 03/21/2024]
Abstract
Morphogenesis during development and regeneration requires cells to communicate and cooperate toward the construction of complex anatomical structures. One important set of mechanisms for coordinating growth and form occurs via developmental bioelectricity-the dynamics of cellular networks driving changes of resting membrane potential which interface with transcriptional and biomechanical downstream cascades. While many molecular details have been elucidated about the instructive processes mediated by ion channel-dependent signaling outside of the nervous system, future advances in regenerative medicine and bioengineering require the understanding of tissue, organ, or whole body-level properties. A key aspect of bioelectric networks is their robustness, which can drive correct, invariant patterning cues despite changing cell number and anatomical configuration of the underlying tissue network. Here, we computationally analyze the minimal models of bioelectric networks and use the example of the regenerating planarian flatworm, to reveal important system-level aspects of bioelectrically derived patterns. These analyses promote an understanding of the robustness of circuits controlling regeneration and suggest design properties that can be exploited for synthetic bioengineering.
Collapse
Affiliation(s)
- Joel Grodstein
- Department of Electrical and Computer Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | | |
Collapse
|
24
|
Electrophysiological engineering of heart-derived cells with calcium-dependent potassium channels improves cell therapy efficacy for cardioprotection. Nat Commun 2021; 12:4963. [PMID: 34400625 PMCID: PMC8368210 DOI: 10.1038/s41467-021-25180-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/21/2021] [Indexed: 12/30/2022] Open
Abstract
We have shown that calcium-activated potassium (KCa)-channels regulate fundamental progenitor-cell functions, including proliferation, but their contribution to cell-therapy effectiveness is unknown. Here, we test the participation of KCa-channels in human heart explant-derived cell (EDC) physiology and therapeutic potential. TRAM34-sensitive KCa3.1-channels, encoded by the KCNN4 gene, are exclusively expressed in therapeutically bioactive EDC subfractions and maintain a strongly polarized resting potential; whereas therapeutically inert EDCs lack KCa3.1 channels and exhibit depolarized resting potentials. Somatic gene transfer of KCNN4 results in membrane hyperpolarization and increases intracellular [Ca2+], which boosts cell-proliferation and the production of pro-healing cytokines/nanoparticles. Intramyocardial injection of EDCs after KCNN4-gene overexpression markedly increases the salutary effects of EDCs on cardiac function, viable myocardium and peri-infarct neovascularization in a well-established murine model of ischemic cardiomyopathy. Thus, electrophysiological engineering provides a potentially valuable strategy to improve the therapeutic value of progenitor cells for cardioprotection and possibly other indications. Strategies to improve the function of damaged hearts with progenitor cells have stalled. Here, the authors show that gene transfer of a calcium-dependent potassium channel enhances the functional properties and ability of explant-derived cells to improve heart function after a heart attack.
Collapse
|
25
|
A bioelectric model of carcinogenesis, including propagation of cell membrane depolarization and reversal therapies. Sci Rep 2021; 11:13607. [PMID: 34193902 PMCID: PMC8245601 DOI: 10.1038/s41598-021-92951-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/19/2021] [Indexed: 12/26/2022] Open
Abstract
As the main theory of carcinogenesis, the Somatic Mutation Theory, increasingly presents difficulties to explain some experimental observations, different theories are being proposed. A major alternative approach is the Tissue Organization Field Theory, which explains cancer origin as a tissue regulation disease instead of having a mainly cellular origin. This work fits in the latter hypothesis, proposing the bioelectric field, in particular the cell membrane polarization state, and ionic exchange through ion channels and gap junctions, as an important mechanism of cell communication and tissue organization and regulation. Taking into account recent experimental results and proposed bioelectric models, a computational model of cancer initiation was developed, including the propagation of a cell depolarization wave in the tissue under consideration. Cell depolarization leads to a change in its state, with the activation and deactivation of several regulation pathways, increasing cell proliferation and motility, changing its epigenetic state to a more stem cell-like behavior without the requirement of genomic mutation. The intercellular communication via gap junctions leads, in certain circumstances, to a bioelectric state propagation to neighbor cells, in a chain-like reaction, till an electric discontinuity is reached. However, this is a reversible process, and it was shown experimentally that, by implementing a therapy targeted on cell ion exchange channels, it is possible to reverse the state and repolarize cells. This mechanism can be an important alternative way in cancer prevention, diagnosis and therapy, and new experiments are proposed to test the presented hypothesis.
Collapse
|
26
|
Chou SM, Li KX, Huang MY, Chen C, Lin King YH, Li GG, Zhou W, Teo CF, Jan YN, Jan LY, Yang SB. Kv1.1 channels regulate early postnatal neurogenesis in mouse hippocampus via the TrkB signaling pathway. eLife 2021; 10:e58779. [PMID: 34018923 PMCID: PMC8208815 DOI: 10.7554/elife.58779] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
In the postnatal brain, neurogenesis occurs only within a few regions, such as the hippocampal sub-granular zone (SGZ). Postnatal neurogenesis is tightly regulated by factors that balance stem cell renewal with differentiation, and it gives rise to neurons that participate in learning and memory formation. The Kv1.1 channel, a voltage-gated potassium channel, was previously shown to suppress postnatal neurogenesis in the SGZ in a cell-autonomous manner. In this study, we have clarified the physiological and molecular mechanisms underlying Kv1.1-dependent postnatal neurogenesis. First, we discovered that the membrane potential of neural progenitor cells is highly dynamic during development. We further established a multinomial logistic regression model for cell-type classification based on the biophysical characteristics and corresponding cell markers. We found that the loss of Kv1.1 channel activity causes significant depolarization of type 2b neural progenitor cells. This depolarization is associated with increased tropomyosin receptor kinase B (TrkB) signaling and proliferation of neural progenitor cells; suppressing TrkB signaling reduces the extent of postnatal neurogenesis. Thus, our study defines the role of the Kv1.1 potassium channel in regulating the proliferation of postnatal neural progenitor cells in mouse hippocampus.
Collapse
Affiliation(s)
- Shu-Min Chou
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Ke-Xin Li
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | | | - Chao Chen
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Yuan-Hung Lin King
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Neuroscience Graduate Program, University of California, San FranciscoSan FranciscoUnited States
| | | | - Wei Zhou
- Department of Anesthesia and Perioperative Care, University of California, San FranciscoSan FranciscoUnited States
| | - Chin Fen Teo
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Lily Yeh Jan
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Neuroscience Program of Academia Sinica, Academia SinicaTaipeiTaiwan
| |
Collapse
|
27
|
Davidian D, Ziman B, Escobar AL, Oviedo NJ. Direct Current Electric Stimulation Alters the Frequency and the Distribution of Mitotic Cells in Planarians. Bioelectricity 2021; 3:77-91. [PMID: 34476379 DOI: 10.1089/bioe.2020.0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background: The use of direct current electric stimulation (DCS) is an effective strategy to treat disease and enhance body functionality. Thus, treatment with DCS is an attractive biomedical alternative, but the molecular underpinnings remain mostly unknown. The lack of experimental models to dissect the effects of DCS from molecular to organismal levels is an important caveat. Here, we introduce the planarian flatworm Schmidtea mediterranea as a tractable organism for in vivo studies of DCS. We developed an experimental method that facilitates the application of direct current electrical stimulation to the whole planarian body (pDCS). Materials and Methods: Planarian immobilization was achieved by combining treatment with anesthesia, agar embedding, and low temperature via a dedicated thermoelectric cooling unit. Electric currents for pDCS were delivered using pulled glass microelectrodes. The electric potential was supplied through a constant voltage power supply. pDCS was administered up to six hours, and behavioral and molecular effects were measured by using video recordings, immunohistochemistry, and gene expression analysis. Results: The behavioral immobilization effects are reversible, and pDCS resulted in a redistribution of mitotic cells along the mediolateral axis of the planarian body. The pDCS effects were dependent on the polarity of the electric field, which led to either increase in reductions in mitotic densities associated with the time of pDCS. The changes in mitotic cells were consistent with apparent redistribution in gene expression of the stem cell marker smedwi-1. Conclusion: The immobilization technique presented in this work facilitates studies aimed at dissecting the effects of exogenous electric stimulation in the adult body. Treatment with DCS can be administered for varying times, and the consequences evaluated at different levels, including animal behavior, cellular and transcriptional changes. Indeed, treatment with pDCS can alter cellular and transcriptional parameters depending on the polarity of the electric field and duration of the exposure.
Collapse
Affiliation(s)
- Devon Davidian
- Department of Molecular & Cell Biology and University of California Merced, Merced, California, USA
| | - Benjamin Ziman
- Department of Molecular & Cell Biology and University of California Merced, Merced, California, USA
| | - Ariel L Escobar
- Department of Bioengineering, University of California Merced, Merced, California, USA
| | - Néstor J Oviedo
- Department of Molecular & Cell Biology and University of California Merced, Merced, California, USA
| |
Collapse
|
28
|
Gelmi A, Schutt CE. Stimuli-Responsive Biomaterials: Scaffolds for Stem Cell Control. Adv Healthc Mater 2021; 10:e2001125. [PMID: 32996270 PMCID: PMC11468740 DOI: 10.1002/adhm.202001125] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/18/2020] [Indexed: 12/28/2022]
Abstract
Stem cell fate is closely intertwined with microenvironmental and endogenous cues within the body. Recapitulating this dynamic environment ex vivo can be achieved through engineered biomaterials which can respond to exogenous stimulation (including light, electrical stimulation, ultrasound, and magnetic fields) to deliver temporal and spatial cues to stem cells. These stimuli-responsive biomaterials can be integrated into scaffolds to investigate stem cell response in vitro and in vivo, and offer many pathways of cellular manipulation: biochemical cues, scaffold property changes, drug release, mechanical stress, and electrical signaling. The aim of this review is to assess and discuss the current state of exogenous stimuli-responsive biomaterials, and their application in multipotent stem cell control. Future perspectives in utilizing these biomaterials for personalized tissue engineering and directing organoid models are also discussed.
Collapse
Affiliation(s)
- Amy Gelmi
- School of ScienceCollege of Science, Engineering and HealthRMIT UniversityMelbourneVIC3001Australia
| | - Carolyn E. Schutt
- Department of Biomedical EngineeringKnight Cancer Institute Cancer Early Detection Advanced Research Center (CEDAR)Oregon Health and Science UniversityPortlandOR97201USA
| |
Collapse
|
29
|
Kamaldinov T, Hahn MS. Dual Bioelectrical Assessment of Human Mesenchymal Stem Cells Using Plasma and Mitochondrial Membrane Potentiometric Probes. Bioelectricity 2020; 2:238-250. [PMID: 34476356 DOI: 10.1089/bioe.2020.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Bioelectrical properties are known to impact stem cell fate, state, and function. However, assays that measure bioelectrical properties are generally limited to the plasma membrane potential. In this study, we propose an assay to simultaneously assess cell plasma membrane and mitochondrial membrane potentials. Materials and Methods: Mesenchymal stem cell (MSC) plasma and mitochondrial membrane potentials were measured using flow cytometry and a combination of tetramethylrhodamine, methyl ester (TMRM), and bis-(1,3-dibutylbarbituric acid)trimethine oxonol (DiBAC) dyes. We investigated the shifts in the bioelectrical phenotype of MSCs due to extended culture in vitro, activation with interferon-gamma (IFN-γ), and aggregate conditions. Results: MSCs subjected to extended culture in vitro acquired plasma and mitochondrial membrane potentials consistent with a hyperpolarized bioelectrical phenotype. Activation with IFN-γ shifted MSCs toward a state associated with increased levels of both DiBAC and TMRM. MSCs in aggregate conditions were associated with a decrease in TMRM levels, indicating mitochondrial depolarization. Conclusions: Our proposed assay described distinct MSC bioelectrical transitions due to extended in vitro culture, exposure to an inflammatory cytokine, and culture under aggregate conditions. Overall, our assay enables a more complete characterization of MSC bioelectrical properties within a single experiment, and its relative simplicity enables researchers to apply it in variety of settings.
Collapse
Affiliation(s)
- Timothy Kamaldinov
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| |
Collapse
|
30
|
Schofield Z, Meloni GN, Tran P, Zerfass C, Sena G, Hayashi Y, Grant M, Contera SA, Minteer SD, Kim M, Prindle A, Rocha P, Djamgoz MBA, Pilizota T, Unwin PR, Asally M, Soyer OS. Bioelectrical understanding and engineering of cell biology. J R Soc Interface 2020; 17:20200013. [PMID: 32429828 PMCID: PMC7276535 DOI: 10.1098/rsif.2020.0013] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/17/2020] [Indexed: 02/07/2023] Open
Abstract
The last five decades of molecular and systems biology research have provided unprecedented insights into the molecular and genetic basis of many cellular processes. Despite these insights, however, it is arguable that there is still only limited predictive understanding of cell behaviours. In particular, the basis of heterogeneity in single-cell behaviour and the initiation of many different metabolic, transcriptional or mechanical responses to environmental stimuli remain largely unexplained. To go beyond the status quo, the understanding of cell behaviours emerging from molecular genetics must be complemented with physical and physiological ones, focusing on the intracellular and extracellular conditions within and around cells. Here, we argue that such a combination of genetics, physics and physiology can be grounded on a bioelectrical conceptualization of cells. We motivate the reasoning behind such a proposal and describe examples where a bioelectrical view has been shown to, or can, provide predictive biological understanding. In addition, we discuss how this view opens up novel ways to control cell behaviours by electrical and electrochemical means, setting the stage for the emergence of bioelectrical engineering.
Collapse
Affiliation(s)
- Zoe Schofield
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Gabriel N. Meloni
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Peter Tran
- Department of Chemical and Biological Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Christian Zerfass
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Giovanni Sena
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Yoshikatsu Hayashi
- Department of Biomedical Engineering, School of Biological Sciences, University of Reading, Reading RG6 6AH, UK
| | - Murray Grant
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Sonia A. Contera
- Clarendon Laboratory, Physics Department, University of Oxford, Parks Road, Oxford OX1 3PU, UK
| | - Shelley D. Minteer
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, USA
| | - Minsu Kim
- Department of Physics, Emory University, Atlanta, GA 30322, USA
| | - Arthur Prindle
- Department of Chemical and Biological Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Paulo Rocha
- Centre for Biosensors, Bioelectronics and Biodevices (C3Bio), Department of Electronic and Electrical Engineering, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Mustafa B. A. Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Teuta Pilizota
- Systems and Synthetic Biology Centre and School of Biological Sciences, University of Edinburgh, Alexander Crum Brown Road, Edinburgh EH9 3FF, UK
| | - Patrick R. Unwin
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK
| | - Munehiro Asally
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Orkun S. Soyer
- Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry CV4 7AL, UK
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
31
|
Kamaldinov T, Erndt-Marino J, Levin M, Kaplan DL, Hahn MS. Assessment of Enrichment of Human Mesenchymal Stem Cells Based on Plasma and Mitochondrial Membrane Potentials. Bioelectricity 2020; 2:21-32. [PMID: 32292894 DOI: 10.1089/bioe.2019.0024] [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] [Indexed: 02/06/2023] Open
Abstract
Background: Human mesenchymal stem cells (hMSCs) are utilized preclinically and clinically as a candidate cell therapy for a wide range of inflammatory and degenerative diseases. Despite promising results in early clinical trials, consistent outcomes with hMSC-based therapies have proven elusive in many of these applications. In this work, we attempt to address this limitation through the design of a stem cell therapy to enrich hMSCs for desired electrical and ionic properties with enhanced stemness and immunomodulatory/regenerative capacity. Materials and Methods: In this study, we sought to develop initial protocols to achieve electrically enriched hMSCs (EE-hMSCs) with distinct electrical states and assess the potential relationship with respect to hMSC state and function. We sorted hMSCs based on fluorescence intensity of tetramethylrhodamine ethyl ester (TMRE) and investigated phenotypic differences between the sorted populations. Results: Subpopulations of EE-hMSCs exhibit differential expression of genes associated with senescence, stemness, immunomodulation, and autophagy. EE-hMSCs with low levels of TMRE, indicative of depolarized membrane potential, have reduced mRNA expression of senescence-associated markers, and increased mRNA expression of autophagy and immunomodulatory markers relative to EE-hMSCs with high levels of TMRE (hyperpolarized). Conclusions : This work suggests that the utilization of EE-hMSCs may provide a novel strategy for cell therapies, enabling live cell enrichment for distinct phenotypes that can be exploited for different therapeutic outcomes.
Collapse
Affiliation(s)
- Timothy Kamaldinov
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Josh Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York.,Department of Biomedical Engineering, Tufts University, Medford, Massachusetts.,Allen Discovery Center at Tufts University, Department of Biology, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology, Tufts University, Medford, Massachusetts
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts.,Allen Discovery Center at Tufts University, Department of Biology, Tufts University, Medford, Massachusetts
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
32
|
Erndt-Marino J, Yeisley DJ, Chen H, Levin M, Kaplan DL, Hahn MS. Interferon-Gamma Stimulated Murine Macrophages In Vitro: Impact of Ionic Composition and Osmolarity and Therapeutic Implications. Bioelectricity 2020; 2:48-58. [PMID: 32292895 PMCID: PMC7107958 DOI: 10.1089/bioe.2019.0032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Injections of osmolytes are promising immunomodulatory treatments for medical benefit, although the rationale and underlying mechanisms are often lacking. The goals of the present study were twofold: (1) to clarify the anti-inflammatory role of the potassium ion and (2) to begin to decouple the effects that ionic strength, ionic species, and osmolarity have on macrophage biology. Materials and Methods: RAW 264.7 murine macrophages were encapsulated in three-dimensional, poly(ethylene glycol) diacrylate hydrogels and activated with interferon-gamma to yield M(IFN). Gene and protein profiles were made of M(IFN) exposed to different hyperosmolar treatments (80 mM potassium gluconate, 80 mM sodium gluconate, and 160 mM sucrose). Results: Relative to M(IFN), all hyperosmolar treatments suppressed expression of pro-inflammatory markers (nitric oxide synthase-2 [NOS-2], tumor necrosis factor-alpha, monocyte chemoattractant protein-1 [MCP-1]) and increased messenger RNA (mRNA) expression of the pleiotropic and angiogenic markers interleukin-6 (IL-6) and vascular endothelial growth factor-A (VEGF), respectively. Ionic osmolytes also demonstrated a greater level of change compared to the nonionic treatments, with mRNA levels of IL-6 the most significantly affected. M(IFN) exposed to K+ exhibited the lowest levels of NOS-2 and MCP-1, and this ion limited IL-6 release induced by osmolarity. Conclusion: Cumulatively, these data suggest that osmolyte composition, ionic strength, and osmolarity are all parameters that can influence therapeutic outcomes. Future work is necessary to further decouple and mechanistically understand the influence that these biophysical parameters have on cell biology, including their impact on other macrophage functions, intracellular osmolyte composition, and cellular and organellular membrane potentials.
Collapse
Affiliation(s)
- Joshua Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Department of Biology, Allen Discovery Center at Tufts University, Tufts University, Medford, Massachusetts
| | - Daniel J. Yeisley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Hongyu Chen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Michael Levin
- Department of Biology, Allen Discovery Center at Tufts University, Tufts University, Medford, Massachusetts
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Department of Biology, Allen Discovery Center at Tufts University, Tufts University, Medford, Massachusetts
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
33
|
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
|
34
|
Grasman JM, Williams MD, Razis CG, Bonzanni M, Golding AS, Cairns DM, Levin M, Kaplan DL. Hyperosmolar potassium inhibits myofibroblast conversion and reduces scar tissue formation. ACS Biomater Sci Eng 2019; 5:5327-5336. [PMID: 32440531 PMCID: PMC7241611 DOI: 10.1021/acsbiomaterials.9b00810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Scar formation is a natural result of almost all wound healing in adult mammals. Unfortunately, scarring disrupts normal tissue function and can cause significant physical and psychological distress. In addition to improving surgical techniques to limit scar formation, several therapies are under development towards the same goal. Many of these treatments aim to disrupt transforming growth factor β1 (TGFβ1) signaling, as this is a critical control point for fibroblast differentiation into myofibroblasts; a contractile cell that organizes synthesized collagen fibrils into scar tissue. The present study aimed to examine the role of hyperosmolar potassium gluconate (KGluc) on fibroblast function in skin repair. KGluc was first determined to negatively regulate fibroblast proliferation, metabolism, and migration in a dose-dependent manner in vitro. Increasing concentrations of KGluc also inhibited differentiation into myofibroblasts, suggesting that local KGluc treatment might reduce fibrosis. KGluc delivery was confirmed via loading into collagen hydrogels and used to treat a full thickness skin wound in mice. KGluc qualitatively slowed initial closure of the wounds and resulted in tissue that more closely resembled mature, healthy skin (epidermal thickness and dermal-epidermal morphology) when compared to unloaded collagen hydrogels. KGluc treatment significantly reduced the number of myofibroblasts within the dermis while upregulated blood vessel density with respect to unloaded hydrogels, likely a result of disruption of TGFβ1 signaling. Taken together, these data demonstrate the effectiveness of KGluc treatment on skin wound healing and suggest that this may be an efficient treatment to limit scar formation.
Collapse
Affiliation(s)
- Jonathan M. Grasman
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Marisa D. Williams
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Constantine G. Razis
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Mattia Bonzanni
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155
| | - Anne S. Golding
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
| | - Dana M. Cairns
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Michael Levin
- Department of Biology, Tufts University, Medford, Massachusetts 02155
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155
| |
Collapse
|
35
|
Abstract
As the leading cause of death in cancer, there is an urgent need to develop treatments to target the dissemination of primary tumor cells to secondary organs, known as metastasis. Bioelectric signaling has emerged in the last century as an important controller of cell growth, and with the development of current molecular tools we are now beginning to identify its role in driving cell migration and metastasis in a variety of cancer types. This review summarizes the currently available research for bioelectric signaling in solid tumor metastasis. We review the steps of metastasis and discuss how these can be controlled by bioelectric cues at the level of a cell, a population of cells, and the tissue. The role of ion channel, pump, and exchanger activity and ion flux is discussed, along with the importance of the membrane potential and the relationship between ion flux and membrane potential. We also provide an overview of the evidence for control of metastasis by external electric fields (EFs) and draw from examples in embryogenesis and regeneration to discuss the implications for endogenous EFs. By increasing our understanding of the dynamic properties of bioelectric signaling, we can develop new strategies that target metastasis to be translated into the clinic.
Collapse
Affiliation(s)
- Samantha L. Payne
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
36
|
Cervera J, Manzanares JA, Mafe S, Levin M. Synchronization of Bioelectric Oscillations in Networks of Nonexcitable Cells: From Single-Cell to Multicellular States. J Phys Chem B 2019; 123:3924-3934. [PMID: 31003574 DOI: 10.1021/acs.jpcb.9b01717] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biological networks use collective oscillations for information processing tasks. In particular, oscillatory membrane potentials have been observed in nonexcitable cells and bacterial communities where specific ion channel proteins contribute to the bioelectric coordination of large populations. We aim at describing theoretically the oscillatory spatiotemporal patterns that emerge at the multicellular level from the single-cell bioelectric dynamics. To this end, we focus on two key questions: (i) What single-cell properties are relevant to multicellular behavior? (ii) What properties defined at the multicellular level can allow an external control of the bioelectric dynamics? In particular, we explore the interplay between transcriptional and translational dynamics and membrane potential dynamics in a model multicellular ensemble, describe the spatiotemporal patterns that arise when the average electric potential allows groups of cells to act as a coordinated multicellular patch, and characterize the resulting synchronization phenomena. The simulations concern bioelectric networks and collective communication across different scales based on oscillatory and synchronization phenomena, thus shedding light on the physiological dynamics of a wide range of endogenous contexts across embryogenesis and regeneration.
Collapse
Affiliation(s)
- Javier Cervera
- Departament de Termodinàmica, Facultat de Física , Universitat de València , E-46100 Burjassot , Spain
| | - José Antonio Manzanares
- Departament de Termodinàmica, Facultat de Física , Universitat de València , E-46100 Burjassot , Spain
| | - Salvador Mafe
- Departament de Termodinàmica, Facultat de Física , Universitat de València , E-46100 Burjassot , Spain
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology , Tufts University Medford , Massachusetts 02155-4243 , United States
| |
Collapse
|
37
|
Erndt-Marino J, Trinkle E, Hahn MS. Hyperosmolar Potassium (K +) Treatment Suppresses Osteoarthritic Chondrocyte Catabolic and Inflammatory Protein Production in a 3-Dimensional In Vitro Model. Cartilage 2019; 10:186-195. [PMID: 28992763 PMCID: PMC6425543 DOI: 10.1177/1947603517734028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE The main goal of this study was to provide a proof-of-concept demonstrating that hyperosmolar K+ solutions can limit production of catabolic and inflammatory mediators in human osteoarthritic chondrocytes (OACs). METHODS A 3-dimensional in vitro model with poly(ethylene glycol) diacrylate (PEGDA) hydrogels was used. Catabolic and pro-inflammatory protein production from encapsulated OACs was assessed following culture for 1 or 7 days in the presence or absence of 80 mM K+ gluconate, 80 mM sodium (Na+) gluconate, or 160 mM sucrose, each added to culture media (final osmolarity ~490 mOsm). RESULTS Relative to untreated controls, OACs treated with hyperosmolar (80 mM Na+ gluconate or 160 mM sucrose) solutions produced lower levels of catabolic and inflammatory mediators in a marker- and time-dependent manner (i.e., MMP-9 after 1 day; MCP-1 after 7 days ( P ≤ 0.015)). In contrast, OAC treatment with 80 mM K+ gluconate reduced catabolic and inflammatory mediators to a greater extent (both the number of markers and degree of suppression) relative to untreated, Na+ gluconate, or sucrose controls (i.e., MMP-3, -9, -13, TIMP-1, MCP-1, and IL-8 after 1 day; MMP-1, -3, -9, -13, TIMP-1, MCP-1, and IL-8 after 7 days ( P ≤ 0.029). CONCLUSIONS Hyperosmolar K+ solutions are capable of attenuating protein production of catabolic and inflammatory OA markers, providing the proof-of-concept needed for further development of a K+-based intra-articular injection for OA treatment. Moreover, K+ performed significantly better than Na+- or sucrose-based solutions, supporting the application of K+ toward improving irrigation solutions for joint surgery.
Collapse
Affiliation(s)
- Josh Erndt-Marino
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Erik Trinkle
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute, Troy, NY, USA,Mariah S. Hahn, Department of Biomedical
Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180,
USA.
| |
Collapse
|
38
|
Tuszynski J, Tilli TM, Levin M. Ion Channel and Neurotransmitter Modulators as Electroceutical Approaches to the Control of Cancer. Curr Pharm Des 2019; 23:4827-4841. [PMID: 28554310 PMCID: PMC6340161 DOI: 10.2174/1381612823666170530105837] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 11/22/2022]
Abstract
The activities of individual cells must be tightly coordinated in order to build and maintain complex 3-dimensional body structures during embryogenesis and regeneration. Thus, one way to view cancer is within systems biology as a network disorder affecting the ability of cells to properly interact with a morphodynamic field of instructive signals that keeps proliferation and migration orchestrated toward the anatomical needs of the host or-ganism. One layer of this set of instructive microenvironmental cues is bioelectrical. Voltage gradients among all somatic cells (not just excitable nerve and muscle) control cell behavior, and the ionic coupling of cells into networks via electrochemical synapses allows them to implement tissue-level patterning decisions. These gradients have been increasingly impli-cated in the induction and suppression of tumorigenesis and metastasis, in the emerging links between developmental bioelectricity to the cancer problem. Consistent with the well-known role of neurotransmitter molecules in transducing electrical activity to downstream cascades in the brain, serotonergic signaling has likewise been implicated in cancer. Here, we review these recent data and propose new approaches for manipulating bioelectric and neurotransmitter pathways in cancer biology based on a bioelectric view of cancer. To sup-port this methodology, we present new data on the effects of the SSRI Prozac and its analog (ZINC ID = ZINC06811610) on survival of both cancer (MCF7) and normal (MCF10A) breast cells exposed to these compounds. We found an IC50 concentration (25 μM for Pro-zac and 100 μM for the Prozac analog) at which these compounds inhibited tumor cell sur-vival and proliferation. Additionally, at these concentrations, we did not observe alterations in a non-tumoral cell line. This constitutes a proof-of-concept demonstration for our hy-pothesis that the use of both existing and novel drugs as electroceuticals could serve as an alternative to highly toxic chemotherapy strategies replacing or augmenting them with less toxic alternatives. We believe this new approach forms an exciting roadmap for future bio-medical advances.
Collapse
Affiliation(s)
- Jack Tuszynski
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta. Canada
| | - Tatiana M Tilli
- Laboratory of Biological System Modeling, National Institute for Science and Technology on Innovation in Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro. Brazil
| | - Michael Levin
- Biology Department, and Allen Discovery Center, Tufts University, Medford, MA, 02155. United States
| |
Collapse
|
39
|
Sundelacruz S, Moody AT, Levin M, Kaplan DL. Membrane Potential Depolarization Alters Calcium Flux and Phosphate Signaling During Osteogenic Differentiation of Human Mesenchymal Stem Cells. Bioelectricity 2019; 1:56-66. [PMID: 32292891 PMCID: PMC6524654 DOI: 10.1089/bioe.2018.0005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Membrane potential (Vmem) changes accompany important events in embryonic development and organ regeneration. Recent studies have pointed to its function as a potent regulator of cell proliferation, differentiation, migration, and tissue regeneration. We have previously reported that Vmem depolarization and hyperpolarization control the osteogenic (OS) differentiation potential of human mesenchymal stem cells (hMSCs). Materials and Methods: In this study, we sought to understand the mechanism(s) underlying voltage regulation of hMSC differentiation. We investigated the role of calcium and phosphate ion flux in the depolarization response of OS-differentiating hMSCs, as these ions are the two major inorganic components of the bone mineral matrix and are indicative of mature osteoblast function. Results: Our results suggest that inorganic phosphate levels play a larger role than calcium flux in mediating hMSC response to depolarization and that the expression of stanniocalcin 1 (STC1), a protein that regulates calcium and phosphate homeostasis in osteoblasts, is functionally required for the depolarization response during the early stages of differentiation. Conclusion: Depolarization alters hMSC differentiation through a phosphate signaling pathway involving STC1. This study enriches our mechanistic understanding of hMSC response to endogenous voltage cues.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Amy Thurber Moody
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology, Medford, Massachusetts
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
40
|
Abstract
Modern stem cell research has mainly focused on protein expression and transcriptional networks. However, transmembrane voltage gradients generated by ion channels and transporters have demonstrated to be powerful regulators of cellular processes. These physiological cues exert influence on cell behaviors ranging from differentiation and proliferation to migration and polarity. Bioelectric signaling is a fundamental element of living systems and an untapped reservoir for new discoveries. Dissecting these mechanisms will allow for novel methods of controlling cell fate and open up new opportunities in biomedicine. This review focuses on the role of ion channels and the resting membrane potential in the proliferation and differentiation of skeletal muscle progenitor cells. In addition, findings relevant to this topic are presented and potential implications for tissue engineering and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Colin Fennelly
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Inc., Cambridge, Massachusetts
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
41
|
Churchill CDM, Winter P, Tuszynski JA, Levin M. EDEn-Electroceutical Design Environment: Ion Channel Tissue Expression Database with Small Molecule Modulators. iScience 2019; 11:42-56. [PMID: 30590250 PMCID: PMC6308252 DOI: 10.1016/j.isci.2018.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/22/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
The emerging field of bioelectricity has revealed numerous new roles for ion channels beyond the nervous system, which can be exploited for applications in regenerative medicine. Developing such biomedical interventions for birth defects, cancer, traumatic injury, and bioengineering first requires knowledge of ion channel targets expressed in tissues of interest. This information can then be used to select combinations of small molecule inhibitors and/or activators that manipulate the bioelectric state. Here, we provide an overview of electroceutical design environment (EDEn), the first bioinformatic platform that facilitates the design of such therapeutic strategies. This database includes information on ion channels and ion pumps, linked to known chemical modulators and their properties. The database also provides information about the expression levels of the ion channels in over 100 tissue types. The graphical interface allows the user to readily identify chemical entities that can alter the electrical properties of target cells and tissues.
Collapse
Affiliation(s)
| | - Philip Winter
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Jack A Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Michael Levin
- Allen Discovery Center, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155, USA.
| |
Collapse
|
42
|
Tarasov MV, Kotova PD, Bystrova MF, Kabanova NV, Sysoeva VY, Kolesnikov SS. Arachidonic acid hyperpolarizes mesenchymal stromal cells from the human adipose tissue by stimulating TREK1 K + channels. Channels (Austin) 2019; 13:36-47. [PMID: 30661462 PMCID: PMC6380217 DOI: 10.1080/19336950.2019.1565251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The current knowledge of electrogenesis in mesenchymal stromal cells (MSCs) remains scarce. Earlier, we demonstrated that in MSCs from the human adipose tissue, transduction of certain agonists involved the phosphoinositide cascade. Its pivotal effector PLC generates DAG that can regulate ion channels directly or via its derivatives, including arachidonic acid (AA). Here we showed that AA strongly hyperpolarized MSCs by stimulating instantly activating, outwardly rectifying TEA-insensitive K+ channels. Among AA-regulated K+ channels, K2P channels from the TREK subfamily appeared to be an appropriate target. The expression of K2P channels in MSCs was verified by RT-PCR, which revealed TWIK-1, TREK-1, and TASK-5 transcripts. The TREK-1 inhibitor spadin antagonized the electrogenic action of AA, which was simulated by the channel activator BL 1249. This functional evidence suggested that TREK-1 channels mediated AA-dependent hyperpolarization of MSCs. Being mostly silent at rest, TREK-1 negligibly contributed to the “background” K+ current. The dramatic stimulation of TREK-1 channels by AA indicates their involvement in AA-dependent signaling in MSCs.
Collapse
Affiliation(s)
- Michail V Tarasov
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Polina D Kotova
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Marina F Bystrova
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Natalia V Kabanova
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Veronika Yu Sysoeva
- b Department of Biochemistry and Molecular Medicine, Faculty of Basic Medicine , Lomonosov Moscow State University , Moscow , Russia
| | - Stanislav S Kolesnikov
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| |
Collapse
|
43
|
Cervera J, Manzanares JA, Mafe S. Cell-cell bioelectrical interactions and local heterogeneities in genetic networks: a model for the stabilization of single-cell states and multicellular oscillations. Phys Chem Chem Phys 2019; 20:9343-9354. [PMID: 29564429 DOI: 10.1039/c8cp00648b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Genetic networks operate in the presence of local heterogeneities in single-cell transcription and translation rates. Bioelectrical networks and spatio-temporal maps of cell electric potentials can influence multicellular ensembles. Could cell-cell bioelectrical interactions mediated by intercellular gap junctions contribute to the stabilization of multicellular states against local genetic heterogeneities? We theoretically analyze this question on the basis of two well-established experimental facts: (i) the membrane potential is a reliable read-out of the single-cell electrical state and (ii) when the cells are coupled together, their individual cell potentials can be influenced by ensemble-averaged electrical potentials. We propose a minimal biophysical model for the coupling between genetic and bioelectrical networks that associates the local changes occurring in the transcription and translation rates of an ion channel protein with abnormally low (depolarized) cell potentials. We then analyze the conditions under which the depolarization of a small region (patch) in a multicellular ensemble can be reverted by its bioelectrical coupling with the (normally polarized) neighboring cells. We show also that the coupling between genetic and bioelectric networks of non-excitable cells, modulated by average electric potentials at the multicellular ensemble level, can produce oscillatory phenomena. The simulations show the importance of single-cell potentials characteristic of polarized and depolarized states, the relative sizes of the abnormally polarized patch and the rest of the normally polarized ensemble, and intercellular coupling.
Collapse
Affiliation(s)
- Javier Cervera
- Dept. Termodinàmica, Fac. Física, Universitat de València, 46100 Burjassot, Spain.
| | - José A Manzanares
- Dept. Termodinàmica, Fac. Física, Universitat de València, 46100 Burjassot, Spain.
| | - Salvador Mafe
- Dept. Termodinàmica, Fac. Física, Universitat de València, 46100 Burjassot, Spain.
| |
Collapse
|
44
|
Ferenc NN, Levin M. Effects of Ivermectin Exposure on Regeneration of D. dorotocephala Planaria: Exploiting Human-Approved Ion Channel Drugs as Morphoceuticals. Macromol Biosci 2018; 19:e1800237. [PMID: 30485697 DOI: 10.1002/mabi.201800237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/22/2018] [Indexed: 01/19/2023]
Abstract
Transformative applications in regenerative medicine await increased control of processes implementing repair and remodeling of complex living structures. Recent work reveals ion channel drugs as a powerful toolkit for modulating endogenous bioelectric circuits that control growth and form in vivo and in vitro. It is therefore especially important to develop assays in model systems that will enable the testing of these "morphoceuticals"-compounds with predictable effects on anatomical structure. The regenerative planaria are an ideal model system for this purpose. Several studies have shown a role for bioelectric signaling in planarian regeneration, but these have focused on Dugesia japonica and Schmidtea mediterranea. It is not known how the alterations of ion channel activity would affect regeneration in other species of planaria-an important aspect of building robust computational models of bioelectric circuits. Here, the effect of ivermectin (IVM), a chloride channel opener drug commonly used to combat heartworm is tested, on regeneration in a new species of planaria: Dugesia dorotocephala. Exposure to IVM during regeneration results in patterning abnormalities, such as bifurcated tails with partial heads, as well as delayed regeneration. These data extend our understanding of the effects of human-approved ion channel drugs on regenerative processes.
Collapse
Affiliation(s)
- Nina N Ferenc
- Chemistry Department, Westfield High School, Chantilly, VA, 20151, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, 200 Boston Ave., Medford, MA, 02155, USA
| |
Collapse
|
45
|
Cervera J, Meseguer S, Mafe S. Intercellular Connectivity and Multicellular Bioelectric Oscillations in Nonexcitable Cells: A Biophysical Model. ACS OMEGA 2018; 3:13567-13575. [PMID: 30411043 PMCID: PMC6217649 DOI: 10.1021/acsomega.8b01514] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/08/2018] [Indexed: 05/28/2023]
Abstract
Bioelectricity is emerging as a crucial mechanism for signal transmission and processing from the single-cell level to multicellular domains. We explore theoretically the oscillatory dynamics that result from the coupling between the genetic and bioelectric descriptions of nonexcitable cells in multicellular ensembles, connecting the genetic prepatterns defined over the ensemble with the resulting spatio-temporal map of cell potentials. These prepatterns assume the existence of a small patch in the ensemble with locally low values of the genetic rate constants that produce a specific ion channel protein whose conductance promotes the cell-polarized state (inward-rectifying channel). In this way, the short-range interactions of the cells within the patch favor the depolarized membrane potential state, whereas the long-range interaction of the patch with the rest of the ensemble promotes the polarized state. The coupling between the local and long-range bioelectric signals allows a binary control of the patch membrane potentials, and alternating cell polarization and depolarization states can be maintained for optimal windows of the number of cells and the intercellular connectivity in the patch. The oscillatory phenomena emerge when the feedback between the single-cell bioelectric and genetic dynamics is coupled at the multicellular level. In this way, the intercellular connectivity acts as a regulatory mechanism for the bioelectrical oscillations. The simulation results are qualitatively discussed in the context of recent experimental studies.
Collapse
Affiliation(s)
- Javier Cervera
- Departamento
de Termodinàmica, Facultat de Física,
Universitat de València, E-46100 Burjassot, Spain
| | - Salvador Meseguer
- Laboratory
of RNA Modification and Mitochondrial Diseases, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Salvador Mafe
- Departamento
de Termodinàmica, Facultat de Física,
Universitat de València, E-46100 Burjassot, Spain
| |
Collapse
|
46
|
Bioelectrical coupling in multicellular domains regulated by gap junctions: A conceptual approach. Bioelectrochemistry 2018; 123:45-61. [DOI: 10.1016/j.bioelechem.2018.04.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/16/2022]
|
47
|
The Potential for Convergence between Synthetic Biology and Bioelectronics. Cell Syst 2018; 7:231-244. [DOI: 10.1016/j.cels.2018.08.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/30/2018] [Accepted: 08/13/2018] [Indexed: 01/20/2023]
|
48
|
Involvement of AMP-activated Protein Kinase (AMPK) in Regulation of Cell Membrane Potential in a Gastric Cancer Cell Line. Sci Rep 2018; 8:6028. [PMID: 29662080 PMCID: PMC5902619 DOI: 10.1038/s41598-018-24460-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/12/2018] [Indexed: 12/26/2022] Open
Abstract
Membrane potential (Vmem) is a key bioelectric property of non-excitable cells that plays important roles in regulating cell proliferation. However, the regulation of Vmem itself remains largely unexplored. We found that, under nutrient starvation, during which cell division is inhibited, MKN45 gastric cancer cells were in a hyperpolarized state associated with a high intracellular chloride concentration. AMP-activated protein kinase (AMPK) activity increased, and expression of cystic fibrosis transmembrane conductance regulator (CFTR) decreased, in nutrient-starved cells. Furthermore, the increase in intracellular chloride concentration level and Vmem hyperpolarization in nutrient-starved cells was suppressed by inhibition of AMPK activity. Intracellular chloride concentrations and hyperpolarization increased after over-activation of AMPK using the specific activator AICAR or suppression of CFTR activity using specific inhibitor GlyH-101. Under these conditions, proliferation of MKN45 cells was inhibited. These results reveal that AMPK controls the dynamic change in Vmem by regulating CFTR and influencing the intracellular chloride concentration, which in turn influences cell-cycle progression. These findings offer new insights into the mechanisms underlying cell-cycle arrest regulated by AMPK and CFTR.
Collapse
|
49
|
McLaughlin KA, Levin M. Bioelectric signaling in regeneration: Mechanisms of ionic controls of growth and form. Dev Biol 2018; 433:177-189. [PMID: 29291972 PMCID: PMC5753428 DOI: 10.1016/j.ydbio.2017.08.032] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022]
Abstract
The ability to control pattern formation is critical for the both the embryonic development of complex structures as well as for the regeneration/repair of damaged or missing tissues and organs. In addition to chemical gradients and gene regulatory networks, endogenous ion flows are key regulators of cell behavior. Not only do bioelectric cues provide information needed for the initial development of structures, they also enable the robust restoration of normal pattern after injury. In order to expand our basic understanding of morphogenetic processes responsible for the repair of complex anatomy, we need to identify the roles of endogenous voltage gradients, ion flows, and electric fields. In complement to the current focus on molecular genetics, decoding the information transduced by bioelectric cues enhances our knowledge of the dynamic control of growth and pattern formation. Recent advances in science and technology place us in an exciting time to elucidate the interplay between molecular-genetic inputs and important biophysical cues that direct the creation of tissues and organs. Moving forward, these new insights enable additional approaches to direct cell behavior and may result in profound advances in augmentation of regenerative capacity.
Collapse
Affiliation(s)
- Kelly A McLaughlin
- Allen Discovery Center, Department of Biology, Tufts University, 200 Boston Ave., Suite 4700, Medford, MA 02155, United States.
| | - Michael Levin
- Allen Discovery Center, Department of Biology, Tufts University, 200 Boston Ave., Suite 4700, Medford, MA 02155, United States
| |
Collapse
|
50
|
Busse SM, McMillen PT, Levin M. Cross-limb communication during Xenopus hind-limb regenerative response: non-local bioelectric injury signals. Development 2018; 145:dev.164210. [DOI: 10.1242/dev.164210] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/31/2018] [Indexed: 12/29/2022]
Abstract
Regeneration of damaged body-parts requires coordination of size, shape, location, and orientation of tissue with the rest of the body. It is not currently known how far injury sites communicate with the remaining soma during repair, or what information may emanate from the injury site to other regions. We examined the bioelectric properties (resting potential gradients in the epidermis) of Xenopus froglets undergoing hind-limb amputation and observed that the contralateral (un-damaged) limb exhibits apparent depolarization signals immediately after the opposite hind-limb is amputated. The pattern of depolarization matches that of the amputated limb and is correlated to the position and type of injury, revealing that information about damage is available to remote body tissues and is detectable non-invasively in vivo by monitoring of the bioelectric state. These data extend knowledge about the electrophysiology of regenerative response, identify a novel communication process via long-range spread of injury signaling, a phenomenon which we call bioelectric injury mirroring (BIM), and suggests revisions to regenerative medicine and diagnostic strategies focused entirely on the wound site and to the use of contralateral limbs as controls.
Collapse
Affiliation(s)
- Sera M. Busse
- Biology Department and Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Patrick T. McMillen
- Biology Department and Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Biology Department and Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| |
Collapse
|