1
|
Bhanothu V. Investigation of the morphological, cellular, biochemical, and molecular modifications in the BG01V human embryonic stem cell-derived neuronal cells. Tissue Cell 2025; 96:102965. [PMID: 40373613 DOI: 10.1016/j.tice.2025.102965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/17/2025]
Abstract
Changes in the morphology, metabolic activity, intracellular calcium (Ca2 +) transients, expression of topoisomerase-2β (Topo-2β), and senescence of human embryonic stem cells (hESCs)-derived neuronal cells on basic hESC culture media and neuronal differentiation medium at different time intervals is not clear. Hence, we aimed to investigate the morphological, cellular, biochemical, and molecular alterations in the BG01V hESC-derived neuronal cells on basic hESC culture media and neuronal differentiation media at different time intervals. MATERIALS AND METHODS BG01V hESC-derived neuronal cells grown on basic hESC culture media and neuronal differentiation media were evaluated for morphological changes by microscopy, metabolic activity by MTT assay, cell viability by Trypan Blue exclusion assay, cellular activity by estimating the Ca2+ deposits, cellular senescence by senescence-associated beta-galactosidase (SA-β-gal) activity, and level of Topo-2β using Western blotting at different time intervals. RESULTS Contrasting to the BG01V hESCs grown on basic hESC culture media, a notable increase in the neuronal cell-like structures, neuritic outgrowth, and expression of nestin protein on neural induction was observed. Higher levels of Ca2+ deposits, metabolic activity, SA-β-gal activity, and Topo-2β expression in BG01V hESC-derived neuronal cells grown on neuronal differentiation media on day 12 compared to hESCs grown on basic hESC culture media including other days were noted. CONCLUSION This study suggests the increase of calcium salts reflecting the calcium activity at distinct phases of neuronal differentiation, ranging from neural induction to neurite extension. The metabolic and SA-β-gal activity of BG01V hESC-derived neuronal cells may suggest the ongoing biological aging process. Upregulation and activation of Topo-2β upon differentiation induction at the mid-phase suggest the activation of inducible gene loci and downregulation of Topo-2β at a later stage.
Collapse
Affiliation(s)
- Venkanna Bhanothu
- Department of Cell Biology, ICMR-National Institute of Nutrition, Tarnaka, Hyderabad, India; Department of Biotechnology & Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, India.
| |
Collapse
|
2
|
Karunasagara S, Bayarkhangai B, Shim HW, Bae HJ, Lee H, Taghizadeh A, Ji Y, Mandakhbayar N, Kim HS, Hyun J, Kim TJ, Lee JH, Kim HW. Electrically-stimulated cellular and tissue events are coordinated through ion channel-mediated calcium influx and chromatin modifications across the cytosol-nucleus space. Biomaterials 2025; 314:122854. [PMID: 39405824 DOI: 10.1016/j.biomaterials.2024.122854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 11/10/2024]
Abstract
Electrical stimulation (ES) through biomaterials and devices has been implicated in activating diverse cell behaviors while facilitating tissue healing process. Despite its significance in modulating biological events, the mechanisms governing ES-activated cellular phenomena remain largely elusive. Here, we demonstrated that millisecond-pulsed temporal ES profoundly impacted a spectrum of cellular events across the membrane-cytosol-nuclear space. These include activated ion channels, intracellular calcium influx, actomyosin contractility, cell migration and proliferation, and secretome release. Such events were coordinated mainly through ES-activated ion channels and calcium oscillation dynamics. Notably, ES increased the chromatin accessibility of genes, particularly those associated with the ES-activated cellular events, underscoring the significance of epigenetic changes in ES-induced behavioral outcomes. We identified histone acetylation (mediated by histone acetyltransferases), among other chromatin modifications, is key in reshaping the chromatin landscape upon ES. These observations were further validated through experiments involving ex vivo skin tissue samples, including activated ion channels and calcium influx, increased cell proliferation and actomyosin contractility, elevated secretome profile, and more accessible chromatin structure following ES. This work provides novel insights into the mechanisms underlying ES-activated cell and tissue events, ultimately guiding design principles for the development of electrical devices and materials effective for tissue repair and wound healing.
Collapse
Affiliation(s)
- Shanika Karunasagara
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Buuvee Bayarkhangai
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye-Won Shim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Han-Jin Bae
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hwalim Lee
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Yunseong Ji
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea; Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Tae-Jin Kim
- Department of Integrated Biological Science, Pusan National University Pusan, 46241, Republic of Korea; Department of Biological Sciences, Pusan National University Pusan, 46241, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
3
|
Zhang L, Ma M, Li J, Qiao K, Xie Y, Zheng Y. Stimuli-responsive microcarriers and their application in tissue repair: A review of magnetic and electroactive microcarrier. Bioact Mater 2024; 39:147-162. [PMID: 38808158 PMCID: PMC11130597 DOI: 10.1016/j.bioactmat.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/07/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Microcarrier applications have made great advances in tissue engineering in recent years, which can load cells, drugs, and bioactive factors. These microcarriers can be minimally injected into the defect to help reconstruct a good microenvironment for tissue repair. In order to achieve more ideal performance and face more complex tissue damage, an increasing amount of effort has been focused on microcarriers that can actively respond to external stimuli. These microcarriers have the functions of directional movement, targeted enrichment, material release control, and providing signals conducive to tissue repair. Given the high controllability and designability of magnetic and electroactive microcarriers, the research progress of these microcarriers is highlighted in this review. Their structure, function and applications, potential tissue repair mechanisms, and challenges are discussed. In summary, through the design with clinical translation ability, meaningful and comprehensive experimental characterization, and in-depth study and application of tissue repair mechanisms, stimuli-responsive microcarriers have great potential in tissue repair.
Collapse
Affiliation(s)
- LiYang Zhang
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Mengjiao Ma
- Beijing Wanjie Medical Device Co., Ltd, Beijing, China
| | - Junfei Li
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Kun Qiao
- Beijing Gerecov Technology Company Ltd., Beijing, China
| | - Yajie Xie
- Beijing Gerecov Technology Company Ltd., Beijing, China
| | - Yudong Zheng
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing, China
| |
Collapse
|
4
|
Lai YS, Chan TW, Nguyen TMH, Lin TC, Chao YY, Wang CY, Hung LY, Tsai SJ, Chiu WT. Store-operated calcium entry inhibits primary ciliogenesis via the activation of Aurora A. FEBS J 2024; 291:1027-1042. [PMID: 38050648 DOI: 10.1111/febs.17024] [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: 08/24/2023] [Revised: 11/02/2023] [Accepted: 12/04/2023] [Indexed: 12/06/2023]
Abstract
The primary cilium is an antenna-like organelle protruding from the cell surface that can detect physical and chemical stimuli in the extracellular space to activate specific signaling pathways and downstream gene expressions. Calcium ion (Ca2+ ) signaling regulates a wide spectrum of cellular processes, including fertilization, proliferation, differentiation, muscle contraction, migration, and death. This study investigated the effects of the regulation of cytosolic Ca2+ levels on ciliogenesis using chemical, genetic, and optogenetic approaches. We found that ionomycin-induced Ca2+ influx inhibited ciliogenesis and Ca2+ chelator BATPA-AM-induced Ca2+ depletion promoted ciliogenesis. In addition, store-operated Ca2+ entry and the endoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1) negatively regulated ciliogenesis. Moreover, an optogenetic platform was used to create different Ca2+ oscillation patterns by manipulating lighting parameters, including density, frequency, exposure time, and duration. Light-activated Ca2+ -translocating channelrhodopsin (CatCh) is activated by 470-nm blue light to induce Ca2+ influx. Our results show that high-frequency Ca2+ oscillations decrease ciliogenesis. Furthermore, the inhibition of cilia formation induced by Ca2+ may occur via the activation of Aurora kinase A. Cilia not only induce Ca2+ signaling but also regulate cilia formation by Ca2+ signaling.
Collapse
Affiliation(s)
- Yi-Shyun Lai
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ta-Wei Chan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Thi My Hang Nguyen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chien Lin
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ying Chao
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, Taiwan
| | - Liang-Yi Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
5
|
Uzieliene I, Popov A, Vaiciuleviciute R, Kirdaite G, Bernotiene E, Ramanaviciene A. Polypyrrole-based structures for activation of cellular functions under electrical stimulation. Bioelectrochemistry 2024; 155:108585. [PMID: 37847982 DOI: 10.1016/j.bioelechem.2023.108585] [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: 05/05/2023] [Revised: 10/04/2023] [Accepted: 10/08/2023] [Indexed: 10/19/2023]
Abstract
Polypyrrole (Ppy) is an electroconductive polymer used in various applications, including in vitro experiments with cell cultures under electrical stimulation (ES). Ppy can be applied in various forms and most importantly, it is biocompatible with cells. Ppy specifically directs ES to cells, which makes Ppy a potential polymer for the development of novel technologies for targeted tissue regeneration. The high potential of ES in combination with different Ppy-based systems, such as hydrogels, scaffolds, or Ppy-layers is advantageous to stimulate cellular differentiation towards neurogenic, cardiac, muscle, and osteogenic lineages. Different in-house devices and the principles of ES application used to stimulate cellular functions are reviewed and summarized. The focus of this review is to observe the most relevant studies and their in-house techniques regarding the application of Ppy-based materials for the use of bone, neural, cardiac, and muscle tissue regeneration under ES. Different types of Ppy materials, such as Ppy particles, layers/films, membranes, and 3D-shaped synthetic and natural scaffolds, as well as combining Ppy with different active molecules are reviewed.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Anton Popov
- Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; NanoTechnas - Center on Nanotechnology and Materials Sciences, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko g. 24, LT-03225 Vilnius, Lithuania
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Gailute Kirdaite
- Department of Experimental, Preventive and Clinical Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; Faculty of Fundamental Sciences, Vilnius Gediminas Technical University, VilniusTech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Almira Ramanaviciene
- Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; NanoTechnas - Center on Nanotechnology and Materials Sciences, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko g. 24, LT-03225 Vilnius, Lithuania.
| |
Collapse
|
6
|
Mantesso A, Nör JE. Stem cells in clinical dentistry. J Am Dent Assoc 2023; 154:1048-1057. [PMID: 37804275 PMCID: PMC11827052 DOI: 10.1016/j.adaj.2023.08.007] [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: 05/15/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Stem cells are present in most of the tissues in the craniofacial complex and play a major role in tissue homeostasis and repair. These cells are characterized by their capacity to differentiate into multiple cell types and to self-renew to maintain a stem cell pool throughout the life of the tissue. TYPES OF STUDIES REVIEWED The authors discuss original data from experiments and comparative analyses and review articles describing the identification and characterization of stem cells of the oral cavity. RESULTS Every oral tissue except enamel, dentin, and cementum contains stem cells for the entire life span. These stem cells self-renew to maintain a pool of cells that can be activated to replace terminally differentiated cells (for example, odontoblasts) or to enable wound healing (for example, dentin bridge in pulp exposures and healing of periodontal tissues after surgery). In addition, dental stem cells can differentiate into functional blood vessels and nerves. Initial clinical trials have shown that transplanting dental pulp stem cells into disinfected necrotic teeth has allowed for the recovery of tooth vitality and vertical and horizontal root growth in immature teeth with incomplete root formation. PRACTICAL IMPLICATIONS As a consequence of these groundbreaking discoveries, stem cell banks are now offering services for the cryopreservation of dental stem cells. The future use of stem cell-based therapies in the clinic will depend on the collaboration of clinicians and researchers in projects designed to understand whether these treatments are safe, efficacious, and clinically feasible.
Collapse
Affiliation(s)
- Andrea Mantesso
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Jacques E. Nör
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
- Department of Otolaryngology, University of Michigan School of Medicine
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Ann Arbor, Michigan, 48109, USA
| |
Collapse
|
7
|
Chen ST, He SY, Li Y, Gu N, Wen C, Lu J. Metallurgical manipulation of surface Volta potential in bimetals and cell response of human mesenchymal stem cells. BIOMATERIALS ADVANCES 2023; 153:213529. [PMID: 37348184 DOI: 10.1016/j.bioadv.2023.213529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Bioelectricity plays an overriding role in directing cell migration, proliferation, differentiation etc. Tailoring the electro-extracellular environment through metallurgical manipulation could modulate the surrounding cell behaviors. In this study, different electric potential patterns, in terms of Volta potential distribution and gradient, were created on the metallic surface as an electric microenvironment, and their effects on adherent human mesenchymal stem cells were investigated. Periodically and randomly distributed Volta potential pattern, respectively, were generated on the surface through spark plasma sintering of two alternatively stacked dissimilar metals films and of a mixture of metallic powders. Actin cytoskeleton staining demonstrated that the Volta potential pattern strongly affected cell attachment and deformation. The cytoskeletons of cells were observed to elongate along the Volta potential gradient and across the border of adjacent regions with higher and lower potentials. Moreover, the steepest potential gradient resulting from the drastic compositional changes on the periodic borders gave rise to the strongest osteogenic tendency among all the samples. This study suggests that tailoring the Volta potential distribution and gradient of metallic biomaterials via metallurgical manipulation is a promising approach to activate surrounding cells, providing an extra degree of freedom for designing desirable bone-repairing metallic implants.
Collapse
Affiliation(s)
- Shi-Ting Chen
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Si-Yuan He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, PR China.
| | - Yan Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Ning Gu
- Medical School, Nanjing University, Nanjing 210093, PR China
| | - Cuie Wen
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
| | - Jian Lu
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong
| |
Collapse
|
8
|
Gao X, Di X, Li J, Kang Y, Xie W, Sun L, Zhang J. Extracellular ATP-induced calcium oscillations regulating the differentiation of osteoblasts through aerobic oxidation metabolism pathways. J Bone Miner Metab 2023; 41:606-620. [PMID: 37418073 DOI: 10.1007/s00774-023-01449-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023]
Abstract
INTRODUCTION The increase of ATP concentration in the extracellular space represents one of the effective signals that stimulate the physiological activities of cells when the bone is exposed to external mechanical stimulation such as stretching and shear stress force throughout life. However, the effects of ATP on osteoblast differentiation and related mechanisms are not well understood. MATERIALS AND METHODS In this study, the roles of extracellular ATP on osteoblast differentiation, intracellular calcium ([Ca2+]i) levels, metabolomics, and the expression of proteins related to energy metabolism were investigated. RESULTS Our results showed that 100 μM extracellular ATP initiated intracellular calcium ([Ca2+]i) oscillations via the calcium-sensing receptor (P2R) and promoted the differentiation of MC3T3-E1 cells. Metabolomics analysis showed that the differentiation of MC3T3-E1 cells depended on aerobic oxidation, but little glycolysis. Moreover, the differentiation of MC3T3-E1 cells and aerobic oxidation were suppressed with the inhibition of AMP-activated protein kinase (AMPK). CONCLUSION These results indicate that calcium oscillations triggered by extracellular ATP can activate aerobic oxidation through AMPK-related signaling pathways and thus promote osteoblast differentiation.
Collapse
Affiliation(s)
- Xiaohang Gao
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 711049, China
| | - Xiaohui Di
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 711049, China
| | - Jingjing Li
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 711049, China
| | - Yiting Kang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 711049, China
| | - Wenjun Xie
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 711049, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Jianbao Zhang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 711049, China.
| |
Collapse
|
9
|
Moccia F, Brunetti V, Soda T, Faris P, Scarpellino G, Berra-Romani R. Store-Operated Ca 2+ Entry as a Putative Target of Flecainide for the Treatment of Arrhythmogenic Cardiomyopathy. J Clin Med 2023; 12:5295. [PMID: 37629337 PMCID: PMC10455538 DOI: 10.3390/jcm12165295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/04/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic disorder that may lead patients to sudden cell death through the occurrence of ventricular arrhythmias. ACM is characterised by the progressive substitution of cardiomyocytes with fibrofatty scar tissue that predisposes the heart to life-threatening arrhythmic events. Cardiac mesenchymal stromal cells (C-MSCs) contribute to the ACM by differentiating into fibroblasts and adipocytes, thereby supporting aberrant remodelling of the cardiac structure. Flecainide is an Ic antiarrhythmic drug that can be administered in combination with β-adrenergic blockers to treat ACM due to its ability to target both Nav1.5 and type 2 ryanodine receptors (RyR2). However, a recent study showed that flecainide may also prevent fibro-adipogenic differentiation by inhibiting store-operated Ca2+ entry (SOCE) and thereby suppressing spontaneous Ca2+ oscillations in C-MSCs isolated from human ACM patients (ACM C-hMSCs). Herein, we briefly survey ACM pathogenesis and therapies and then recapitulate the main molecular mechanisms targeted by flecainide to mitigate arrhythmic events, including Nav1.5 and RyR2. Subsequently, we describe the role of spontaneous Ca2+ oscillations in determining MSC fate. Next, we discuss recent work showing that spontaneous Ca2+ oscillations in ACM C-hMSCs are accelerated to stimulate their fibro-adipogenic differentiation. Finally, we describe the evidence that flecainide suppresses spontaneous Ca2+ oscillations and fibro-adipogenic differentiation in ACM C-hMSCs by inhibiting constitutive SOCE.
Collapse
Affiliation(s)
- Francesco Moccia
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.B.); (G.S.)
| | - Valentina Brunetti
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.B.); (G.S.)
| | - Teresa Soda
- Department of Health Sciences, University of Magna Graecia, 88100 Catanzaro, Italy;
| | - Pawan Faris
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Giorgia Scarpellino
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.B.); (G.S.)
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla 72410, Mexico;
| |
Collapse
|
10
|
Wang H, Tian J, Jiang Y, Liu S, Zheng J, Li N, Wang G, Dong F, Chen J, Xie Y, Huang Y, Cai X, Wang X, Xiong W, Qi H, Yin L, Wang Y, Sheng X. A 3D biomimetic optoelectronic scaffold repairs cranial defects. SCIENCE ADVANCES 2023; 9:eabq7750. [PMID: 36791200 PMCID: PMC9931229 DOI: 10.1126/sciadv.abq7750] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
Bone fractures and defects pose serious health-related issues on patients. For clinical therapeutics, synthetic scaffolds have been actively explored to promote critical-sized bone regeneration, and electrical stimulations are recognized as an effective auxiliary to facilitate the process. Here, we develop a three-dimensional (3D) biomimetic scaffold integrated with thin-film silicon (Si)-based microstructures. This Si-based hybrid scaffold not only provides a 3D hierarchical structure for guiding cell growth but also regulates cell behaviors via photo-induced electrical signals. Remotely controlled by infrared illumination, these Si structures electrically modulate membrane potentials and intracellular calcium dynamics of stem cells and potentiate cell proliferation and differentiation. In a rodent model, the Si-integrated scaffold demonstrates improved osteogenesis under optical stimulations. Such a wirelessly powered optoelectronic scaffold eliminates tethered electrical implants and fully degrades in a biological environment. The Si-based 3D scaffold combines topographical and optoelectronic stimuli for effective biological modulations, offering broad potential for biomedicine.
Collapse
Affiliation(s)
- Huachun Wang
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
| | - Jingjing Tian
- Department of Medical Science Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yuxi Jiang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing 100082, China
| | - Shuang Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jingchuan Zheng
- School of Materials Science and Engineering, State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Ningyu Li
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing 100082, China
| | - Guiyan Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing 100082, China
| | - Fan Dong
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing 100082, China
| | - Junyu Chen
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
| | - Yang Xie
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
| | - Yunxiang Huang
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
| | - Xue Cai
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
| | - Xiumei Wang
- School of Materials Science and Engineering, State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Wei Xiong
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Hui Qi
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China
| | - Lan Yin
- School of Materials Science and Engineering, State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yuguang Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing 100082, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing 100084, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
11
|
Maione AS, Faris P, Iengo L, Catto V, Bisonni L, Lodola F, Negri S, Casella M, Guarino A, Polvani G, Cerrone M, Tondo C, Pompilio G, Sommariva E, Moccia F. Ca 2+ dysregulation in cardiac stromal cells sustains fibro-adipose remodeling in Arrhythmogenic Cardiomyopathy and can be modulated by flecainide. J Transl Med 2022; 20:522. [PMID: 36371290 PMCID: PMC9652790 DOI: 10.1186/s12967-022-03742-8] [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: 09/07/2022] [Accepted: 10/30/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Cardiac mesenchymal stromal cells (C-MSC) were recently shown to differentiate into adipocytes and myofibroblasts to promote the aberrant remodeling of cardiac tissue that characterizes arrhythmogenic cardiomyopathy (ACM). A calcium (Ca2+) signaling dysfunction, mainly demonstrated in mouse models, is recognized as a mechanism impacting arrhythmic risk in ACM cardiomyocytes. Whether similar mechanisms influence ACM C-MSC fate is still unknown. Thus, we aim to ascertain whether intracellular Ca2+ oscillations and the Ca2+ toolkit are altered in human C-MSC obtained from ACM patients, and to assess their link with C-MSC-specific ACM phenotypes. METHODS AND RESULTS ACM C-MSC show enhanced spontaneous Ca2+ oscillations and concomitant increased Ca2+/Calmodulin dependent kinase II (CaMKII) activation compared to control cells. This is manly linked to a constitutive activation of Store-Operated Ca2+ Entry (SOCE), which leads to enhanced Ca2+ release from the endoplasmic reticulum through inositol-1,4,5-trisphosphate receptors. By targeting the Ca2+ handling machinery or CaMKII activity, we demonstrated a causative link between Ca2+ oscillations and fibro-adipogenic differentiation of ACM C-MSC. Genetic silencing of the desmosomal gene PKP2 mimics the remodelling of the Ca2+ signalling machinery occurring in ACM C-MSC. The anti-arrhythmic drug flecainide inhibits intracellular Ca2+ oscillations and fibro-adipogenic differentiation by selectively targeting SOCE. CONCLUSIONS Altogether, our results extend the knowledge of Ca2+ dysregulation in ACM to the stromal compartment, as an etiologic mechanism of C-MSC-related ACM phenotypes. A new mode of action of flecainide on a novel mechanistic target is unveiled against the fibro-adipose accumulation in ACM.
Collapse
Affiliation(s)
- Angela S Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy.
| | - Pawan Faris
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Lara Iengo
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
| | - Valentina Catto
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Luca Bisonni
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
| | - Francesco Lodola
- Laboratory of Cardiac Cellular Physiology, Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Sharon Negri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Michela Casella
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Cardiology and Arrhythmology Clinic, University Hospital "Umberto I-Salesi-Lancisi", Ancona, Italy
| | - Anna Guarino
- Cardiovascular Tissue Bank of Lombardy, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gianluca Polvani
- Cardiovascular Tissue Bank of Lombardy, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Marina Cerrone
- Medicine, Leon H. Charney Division of Cardiology, Heart Rhythm Center and Cardiovascular Genetics Program, New York University School of Medicine, New York, USA
| | - Claudio Tondo
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dentist Sciences, University of Milano, Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dentist Sciences, University of Milano, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
12
|
Pulsed Electrical Stimulation Affects Osteoblast Adhesion and Calcium Ion Signaling. Cells 2022; 11:cells11172650. [PMID: 36078058 PMCID: PMC9454840 DOI: 10.3390/cells11172650] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
An extensive research field in regenerative medicine is electrical stimulation (ES) and its impact on tissue and cells. The mechanism of action of ES, particularly the role of electrical parameters like intensity, frequency, and duration of the electric field, is not yet fully understood. Human MG-63 osteoblasts were electrically stimulated for 10 min with a commercially available multi-channel system (IonOptix). We generated alternating current (AC) electrical fields with a voltage of 1 or 5 V and frequencies of 7.9 or 20 Hz, respectively. To exclude liquid-mediated effects, we characterized the AC-stimulated culture medium. AC stimulation did not change the medium’s pH, temperature, and oxygen content. The H2O2 level was comparable with the unstimulated samples except at 5 V_7.9 Hz, where a significant increase in H2O2 was found within the first 30 min. Pulsed electrical stimulation was beneficial for the process of attachment and initial adhesion of suspended osteoblasts. At the same time, the intracellular Ca2+ level was enhanced and highest for 20 Hz stimulated cells with 1 and 5 V, respectively. In addition, increased Ca2+ mobilization after an additional trigger (ATP) was detected at these parameters. New knowledge was provided on why electrical stimulation contributes to cell activation in bone tissue regeneration.
Collapse
|
13
|
Celik E, Ercin M, Bolkent S, Gezginci-Oktayoglu S. Metformin induces mitochondrial remodeling and differentiation of pancreatic progenitor cells into beta-cells by a potential mechanism including suppression of the T1R3, PLCβ2, cytoplasmic Ca +2, and AKT. J Physiol Biochem 2022; 78:869-883. [PMID: 35907121 DOI: 10.1007/s13105-022-00910-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022]
Abstract
The main goal of this study was to investigate the molecular changes in pancreatic progenitor cells subject to high glucose, aspartame, and metformin in vitro. This scope of work glucose, aspartame, and metformin were exposed to pancreatic islet derived progenitor cells (PID-PCs) for 10 days. GLUT1's role in beta-cell differentiation was examined by using GLUT1 inhibitor WZB117. Insulin+ cell ratio was measured by flow cytometry; the expression of beta-cell differentiation related genes was shown by RT-PCR; mitochondrial mass, mitochondrial ROS level, cytoplasmic Ca2+, glucose uptake, and metabolite analysis were made fluorometrically and spectrophotometrically; and proteins involved in related molecular pathways were determined by western blotting. Findings showed that glucose or aspartame exposed cells had similar metabolic and gene expression profile to control PID-PCs. Furthermore, relatively few insulin+ cells in aspartame treated cells were determined. Aspartame signal is transmitted through PLCβ2, CAMKK2 and LKB1 in PID-PCs. The most obvious finding of this study is that metformin significantly increased beta-cell differentiation. The mechanism involves suppression of the sweet taste signal's molecules T1R3, PLCβ2, cytoplasmic Ca+2, and AKT in addition to the direct effect of metformin on mitochondria and AMPK, and the energy metabolism of PID-PCs is remodelled in the direction of oxidative phosphorylation. These findings are very important in terms of determining that metformin stimulates the mitochondrial remodeling and the differentiation of PID-PCs to beta-cells and thus it may contribute to the compensation step, which is the first stage of diabetes development.
Collapse
Affiliation(s)
- Ertan Celik
- Molecular Biology Program, Biology Section, Institute of Science, Istanbul University, Istanbul, Turkey
| | - Merve Ercin
- Molecular Biology Program, Biology Section, Institute of Science, Istanbul University, Istanbul, Turkey.,Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey
| | - Sehnaz Bolkent
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey
| | - Selda Gezginci-Oktayoglu
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.
| |
Collapse
|
14
|
Guillot-Ferriols M, Lanceros-Méndez S, Gómez Ribelles JL, Gallego Ferrer G. Electrical stimulation: Effective cue to direct osteogenic differentiation of mesenchymal stem cells? BIOMATERIALS ADVANCES 2022; 138:212918. [PMID: 35913228 DOI: 10.1016/j.bioadv.2022.212918] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/02/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Mesenchymal stem cells (MSCs) play a major role in bone tissue engineering (BTE) thanks to their capacity for osteogenic differentiation and being easily available. In vivo, MSCs are exposed to an electroactive microenvironment in the bone niche, which has piezoelectric properties. The correlation between the electrically active milieu and bone's ability to adapt to mechanical stress and self-regenerate has led to using electrical stimulation (ES) as physical cue to direct MSCs differentiation towards the osteogenic lineage in BTE. This review summarizes the different techniques to electrically stimulate MSCs to induce their osteoblastogenesis in vitro, including general electrical stimulation and substrate mediated stimulation by means of conductive or piezoelectric cell culture supports. Several aspects are covered, including stimulation parameters, treatment times and cell culture media to summarize the best conditions for inducing MSCs osteogenic commitment by electrical stimulation, from a critical point of view. Electrical stimulation activates different signaling pathways, including bone morphogenetic protein (BMP) Smad-dependent or independent, regulated by mitogen activated protein kinases (MAPK), extracellular signal-regulated kinases (ERK) and p38. The roles of voltage gate calcium channels (VGCC) and integrins are also highlighted according to their application technique and parameters, mainly converging in the expression of RUNX2, the master regulator of the osteogenic differentiation pathway. Despite the evident lack of homogeneity in the approaches used, the ever-increasing scientific evidence confirms ES potential as an osteoinductive cue, mimicking aspects of the in vivo microenvironment and moving one step forward to the translation of this approach into clinic.
Collapse
Affiliation(s)
- M Guillot-Ferriols
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain.
| | - S Lanceros-Méndez
- Centre of Physics of Minho and Porto Universities, Universidade do Minho, 4710-058 Braga, Portugal; BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - J L Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - G Gallego Ferrer
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| |
Collapse
|
15
|
Naderi M, Nadri S. Synergistic effect of miR-9 overexpression and electrical induction on differentiation of conjunctiva mesenchymal stem cells into photoreceptor-like cells. Int J Artif Organs 2022; 45:623-630. [PMID: 35658561 DOI: 10.1177/03913988221103285] [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: 11/17/2022]
Abstract
A variety of genes and materials can induce the differentiation of stem cells. The purpose of this work was to investigate the effect of microRNA-9 (miR-9) overexpression and electrical induction on the photoreceptor differentiation of Conjunctiva Mesenchymal Stem Cells (CJMSCs). In this study, an electroconductive scaffold (silk fibroin polymer (SF) and reduced graphene oxide (rGo) nanoparticles) was fabricated by electrospinning method, and its characteristics such as diameter, graphene distribution, compound, conductivity, and toxicity were evaluated by scanning and transmission electron microscopy (SEM and TEM), FTIR, electrochemical impedance spectroscopy, and MTT assay. The cells were transduced by a lentiviral vector carrying miR-9, then electrical induction was implied on mir-9-CJMSCs, cultivated on the fabricated scaffold, and the expressions of neural and photoreceptor marker genes were evaluated by RT-qPCR. A uniform, smooth appearance with lower diameter, uniform distribution of rGo nanoparticles across the fibers, and lower resistance were shown in SF-rGo fibrous scaffold. After electrical stimulation, lower and higher expression of neural marker genes and photoreceptor marker genes (Rhodopsin, PKC) were documented, respectively. Finally, we proposed that the combinational approach of miR-9 overexpression and electrical induction leads CJMSCs to photoreceptor-like cells.
Collapse
Affiliation(s)
- Mahmood Naderi
- Cell-Based Therapies Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samad Nadri
- Department of Medical Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
16
|
Haroutunian GG, Tsaghikian A, Fedorova E, Chaurasia P, Gusella GL, Mosoian A. Electromagnetic Fields Generated by the IteraCoil Device Differentiate Mesenchymal Stem Progenitor Cells Into the Osteogenic Lineage. Bioelectromagnetics 2022; 43:245-256. [PMID: 35391494 PMCID: PMC9325380 DOI: 10.1002/bem.22401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/11/2022] [Accepted: 03/20/2022] [Indexed: 11/09/2022]
Abstract
Rapid advances in mesenchymal stem progenitor cells (MSPCs) have rendered impetus into the area of cell therapy and regenerative medicine. The main promise of future stem cell therapies is their reliance on autologous stem cells derived from adipose tissue, which also includes treatments of bone fractures and degeneration. The effectiveness of different electric devices utilized to reprogram MSPCs toward osteogenic differentiation has provided varying degrees of effectiveness for clinical use. Adipose tissue-derived MSPCs were flow-cytometrically characterized and further differentiated into osteoblasts by culturing either in growth medium with pro-osteogenic supplements or without supplements with alternating electromagnetic field (EMF) generated by IteraCoil. IteraCoil is a multi-solenoid coil with a specific complex geometry that creates a 3D-EMF with desired parameters without directly applying electrodes to the cells and tissues. The flow-cytometric analysis of highly enriched (≥95%) adipose-derived MSPCs (CD34- , CD73+ , CD90+ , and CD105+ ) was utilized for the study. Osteoblasts and chondrocyte differentiations were then assessed by specific staining and quantified using ImageJ (National Institutes of Health). The osteoblastic differentiation of MSPCs cultured in regular medium and exposed to EMF at 0.05 and 1 kHz frequencies was compared with MSPCs cultured in a pro-osteogenic supplemented medium. In this study, we demonstrated that EMF from IteraCoil might have affected the signaling pathways that induce the osteogenic differentiation of human adipose-derived MSPCs in the absence of exogenous osteogenic factors. Therefore, EMF-generated osteogenic differentiation of reprogrammed adipose-derived autologous MSPCs may treat the loss of osteoblasts and osteoporosis and open new avenues for the development of regenerative cellular therapy. © 2022 Bioelectromagnetics Society.
Collapse
Affiliation(s)
| | - Ashot Tsaghikian
- Data Processing and Field Engineering Corp., Glendale, California
| | | | | | | | | |
Collapse
|
17
|
Muneekaew S, Wang MJ, Chen SY. Control of stem cell differentiation by using extrinsic photobiomodulation in conjunction with cell adhesion pattern. Sci Rep 2022; 12:1812. [PMID: 35110659 PMCID: PMC8811059 DOI: 10.1038/s41598-022-05888-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
The induction and direction of stem cell differentiation into needed cell phenotypes is the central pillar of tissue engineering for repairing damaged tissues or organs. Conventionally, a special recipe of chemical factors is formulated to achieve this purpose for each specific target cell type. In this work, it is demonstrated that the combination of extrinsic photobiomodulation and collagen-covered microislands could be used to induce differentiation of Wharton’s jelly mesenchymal stem cells (WJ-MSCs) with the differentiation direction dictated by the specific island topography without use of chemical factors. Both neurogenic differentiation and adipogenic differentiation could be attained with a rate surpassing that using chemical factors. Application of this method to other cell types is possible by utilizing microislands with a pattern tailored particularly for each specific cell type, rendering it a versatile modality for initiating and guiding stem cell differentiation.
Collapse
Affiliation(s)
- Saitong Muneekaew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City, 106, Taiwan
| | - Meng-Jiy Wang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City, 106, Taiwan.
| | - Szu-Yuan Chen
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei City, 106, Taiwan. .,Department of Physics, National Central University, Taoyuan City, 320, Taiwan.
| |
Collapse
|
18
|
Amaroli A, Pasquale C, Zekiy A, Benedicenti S, Marchegiani A, Sabbieti MG, Agas D. Steering the multipotent mesenchymal cells towards an anti-inflammatory and osteogenic bias via photobiomodulation therapy: How to kill two birds with one stone. J Tissue Eng 2022; 13:20417314221110192. [PMID: 35832724 PMCID: PMC9272199 DOI: 10.1177/20417314221110192] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/13/2022] [Indexed: 12/17/2022] Open
Abstract
The bone marrow-derived multipotent mesenchymal cells (MSCs) have captured scientific interest due to their multi-purpose features and clinical applications. The operational dimension of MSCs is not limited to the bone marrow reservoir, which exerts bone-building and niche anabolic tasks; they also meet the needs of quenching inflammation and restoring inflamed tissues. Thus, the range of MSC activities extends to conditions such as neurodegenerative diseases, immune disorders and various forms of osteopenia. Steering these cells towards becoming an effective therapeutic tool has become mandatory. Many laboratories have employed distinct strategies to improve the plasticity and secretome of MSCs. We aimed to present how photobiomodulation therapy (PBM-t) can manipulate MSCs to render them an extraordinary anti-inflammatory and osteogenic instrument. Moreover, we discuss the outcomes of different PBM-t protocols on MSCs, concluding with some perplexities and complexities of PBM-t in vivo but encouraging and feasible in vitro solutions.
Collapse
Affiliation(s)
- Andrea Amaroli
- Department of Surgical and Diagnostic Sciences, University of Genoa, Genoa, Italy.,Department of Orthopedic Dentistry, Faculty of Dentistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Claudio Pasquale
- Department of Surgical and Diagnostic Sciences, University of Genoa, Genoa, Italy
| | - Angelina Zekiy
- Department of Orthopedic Dentistry, Faculty of Dentistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Stefano Benedicenti
- Department of Surgical and Diagnostic Sciences, University of Genoa, Genoa, Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | | | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| |
Collapse
|
19
|
Maltan L, Najjar H, Tiffner A, Derler I. Deciphering Molecular Mechanisms and Intervening in Physiological and Pathophysiological Processes of Ca 2+ Signaling Mechanisms Using Optogenetic Tools. Cells 2021; 10:3340. [PMID: 34943850 PMCID: PMC8699489 DOI: 10.3390/cells10123340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Calcium ion channels are involved in numerous biological functions such as lymphocyte activation, muscle contraction, neurotransmission, excitation, hormone secretion, gene expression, cell migration, memory, and aging. Therefore, their dysfunction can lead to a wide range of cellular abnormalities and, subsequently, to diseases. To date various conventional techniques have provided valuable insights into the roles of Ca2+ signaling. However, their limited spatiotemporal resolution and lack of reversibility pose significant obstacles in the detailed understanding of the structure-function relationship of ion channels. These drawbacks could be partially overcome by the use of optogenetics, which allows for the remote and well-defined manipulation of Ca2+-signaling. Here, we review the various optogenetic tools that have been used to achieve precise control over different Ca2+-permeable ion channels and receptors and associated downstream signaling cascades. We highlight the achievements of optogenetics as well as the still-open questions regarding the resolution of ion channel working mechanisms. In addition, we summarize the successes of optogenetics in manipulating many Ca2+-dependent biological processes both in vitro and in vivo. In summary, optogenetics has significantly advanced our understanding of Ca2+ signaling proteins and the used tools provide an essential basis for potential future therapeutic application.
Collapse
Affiliation(s)
| | | | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (L.M.); (H.N.); (A.T.)
| |
Collapse
|
20
|
McDonough RC, Price C. Targeted Activation of GPCR-Mediated Ca 2+ Signaling Drives Enhanced Cartilage-Like Matrix Formation. Tissue Eng Part A 2021; 28:405-419. [PMID: 34693731 PMCID: PMC9271335 DOI: 10.1089/ten.tea.2021.0078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Intracellular calcium ([Ca2+]i) signaling is a critical regulator of chondrogenesis, chondrocyte differentiation, and cartilage development. Calcium (Ca2+) signaling is known to direct processes that govern chondrocyte gene expression, protein synthesis, cytoskeletal remodeling, and cell fate. Control of chondrocyte/chondroprogenitor Ca2+ signaling has been attempted through mechanical and/or pharmacological activation of endogenous Ca2+ signaling transducers; however, such approaches can lack specificity and/or precision regarding Ca2+ activation mechanisms. Synthetic signaling platforms permitting precise and selective Ca2+ signal transduction can improve dissection of the roles that [Ca2+]i signaling play in chondrocyte behavior. One such platform is the chemogenetic hM3Dq DREADD (designer receptor exclusively activated by designer drugs) that activates [Ca2+]i signaling via the Gαq-PLCβ-IP3-ER pathway upon clozapine N-oxide (CNO) administration. We previously demonstrated hM3Dq's ability to precisely and synthetically initiate robust [Ca2+]i transients and oscillatory [Ca2+]i signaling in chondrocyte-like ATDC5 cells. Here, we investigate the effects that long-term CNO stimulatory culture have on hM3Dq [Ca2+]i signaling dynamics, proliferation, and protein deposition in 2D ATDC5 cultures. Long-term culturing under repeated CNO stimulation modified the temporal dynamics of hM3Dq [Ca2+]i signaling, increased cell proliferation, and enhanced matrix production in a CNO dose- and frequency-dependent manner, and triggered the formation of cell condensations that developed aligned, anisotropic neotissue structures rich in cartilaginous proteoglycans and collagens, all in the absence of differentiation inducers. This study demonstrated Gαq-GPCR-mediated [Ca2+]i signaling involvement in chondroprogenitor proliferation and cartilage-like matrix production, and established hM3Dq as a powerful tool for elucidating the role of GPCR-mediated Ca2+ signaling in chondrogenesis and chondrocyte differentiation.
Collapse
Affiliation(s)
- Ryan C McDonough
- University of Delaware, 5972, Biomedical Engineering, 161 Colburn Lab, Newark, Delaware, United States, 19716-5600;
| | - Christopher Price
- University of Delaware, 5972, Biomedical Engineering, Newark, Delaware, United States;
| |
Collapse
|
21
|
Torre EC, Bicer M, Cottrell GS, Widera D, Tamagnini F. Time-Dependent Reduction of Calcium Oscillations in Adipose-Derived Stem Cells Differentiating towards Adipogenic and Osteogenic Lineage. Biomolecules 2021; 11:biom11101400. [PMID: 34680033 PMCID: PMC8533133 DOI: 10.3390/biom11101400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived mesenchymal stromal cells (ASCs) are multipotent stem cells which can differentiate into various cell types, including osteocytes and adipocytes. Due to their ease of harvesting, multipotency, and low tumorigenicity, they are a prime candidate for the development of novel interventional approaches in regenerative medicine. ASCs exhibit slow, spontaneous Ca2+ oscillations and the manipulation of Ca2+ signalling via electrical stimulation was proposed as a potential route for promoting their differentiation in vivo. However, the effects of differentiation-inducing treatments on spontaneous Ca2+ oscillations in ASCs are not yet fully characterised. In this study, we used 2-photon live Ca2+ imaging to assess the fraction of cells showing spontaneous oscillations and the frequency of the oscillation (measured as interpeak interval—IPI) in ASCs undergoing osteogenic or adipogenic differentiation, using undifferentiated ASCs as controls. The measurements were carried out at 7, 14, and 21 days in vitro (DIV) to assess the effect of time in culture on Ca2+ dynamics. We observed that both time and differentiation treatment are important factors associated with a reduced fraction of cells showing Ca2+ oscillations, paralleled by increased IPI times, in comparison with untreated ASCs. Both adipogenic and osteogenic differentiation resulted in a reduction in Ca2+ dynamics, such as the fraction of cells showing intracellular Ca2+ oscillations and their frequency. Adipogenic differentiation was associated with a more pronounced reduction of Ca2+ dynamics compared to cells differentiating towards the osteogenic fate. Changes in Ca2+ associated oscillations with a specific treatment had already occurred at 7 DIV. Finally, we observed a reduction in Ca2+ dynamics over time in untreated ASCs. These data suggest that adipogenic and osteogenic differentiation cell fates are associated with specific changes in spontaneous Ca2+ dynamics over time. While this observation is interesting and provides useful information to understand the functional correlates of stem cell differentiation, further studies are required to clarify the molecular and mechanistic correlates of these changes. This will allow us to better understand the causal relationship between Ca2+ dynamics and differentiation, potentially leading to the development of novel, more effective interventions for both bone regeneration and control of adipose growth.
Collapse
Affiliation(s)
- Enrico C. Torre
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; (E.C.T.); (M.B.)
- Neuronal and Cellular Physiology Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
- Biomedicine West Wing, International Centre for Life, Times Square, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Mesude Bicer
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; (E.C.T.); (M.B.)
- Department of Bioengineering, Sumer Campus, Abdullah Gül University, Kayseri 38080, Turkey
| | - Graeme S. Cottrell
- Cellular and Molecular Neuroscience, School of Pharmacy, University of Reading, Reading RG6 6LA, UK;
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; (E.C.T.); (M.B.)
- Correspondence: (D.W.); (F.T.)
| | - Francesco Tamagnini
- Neuronal and Cellular Physiology Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
- Correspondence: (D.W.); (F.T.)
| |
Collapse
|
22
|
Primary cilia in hard tissue development and diseases. Front Med 2021; 15:657-678. [PMID: 34515939 DOI: 10.1007/s11684-021-0829-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/13/2020] [Indexed: 10/20/2022]
Abstract
Bone and teeth are hard tissues. Hard tissue diseases have a serious effect on human survival and quality of life. Primary cilia are protrusions on the surfaces of cells. As antennas, they are distributed on the membrane surfaces of almost all mammalian cell types and participate in the development of organs and the maintenance of homeostasis. Mutations in cilium-related genes result in a variety of developmental and even lethal diseases. Patients with multiple ciliary gene mutations present overt changes in the skeletal system, suggesting that primary cilia are involved in hard tissue development and reconstruction. Furthermore, primary cilia act as sensors of external stimuli and regulate bone homeostasis. Specifically, substances are trafficked through primary cilia by intraflagellar transport, which affects key signaling pathways during hard tissue development. In this review, we summarize the roles of primary cilia in long bone development and remodeling from two perspectives: primary cilia signaling and sensory mechanisms. In addition, the cilium-related diseases of hard tissue and the manifestations of mutant cilia in the skeleton and teeth are described. We believe that all the findings will help with the intervention and treatment of related hard tissue genetic diseases.
Collapse
|
23
|
Srirussamee K, Xue R, Mobini S, Cassidy NJ, Cartmell SH. Changes in the extracellular microenvironment and osteogenic responses of mesenchymal stem/stromal cells induced by in vitro direct electrical stimulation. J Tissue Eng 2021; 12:2041731420974147. [PMID: 33643602 PMCID: PMC7894594 DOI: 10.1177/2041731420974147] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022] Open
Abstract
Electrical stimulation (ES) has potential to be an effective tool for bone injury treatment in clinics. However, the therapeutic mechanism associated with ES is still being discussed. This study aims to investigate the initial mechanism of action by characterising the physical and chemical changes in the extracellular environment during ES and correlate them with the responses of mesenchymal stem/stromal cells (MSCs). Computational modelling was used to estimate the electrical potentials relative to the cathode and the current density across the cell monolayer. We showed expression of phosphorylated ERK1/2, c-FOS, c-JUN, and SPP1 mRNAs, as well as the increased metabolic activities of MSCs at different time points. Moreover, the average of 2.5 μM of H2O2 and 34 μg/L of dissolved Pt were measured from the electrically stimulated media (ES media), which also corresponded with the increases in SPP1 mRNA expression and cell metabolic activities. The addition of sodium pyruvate to the ES media as an antioxidant did not alter the SPP1 mRNA expression, but eliminated an increase in cell metabolic activities induced by ES media treatment. These findings suggest that H2O2 was influencing cell metabolic activity, whereas SPP1 mRNA expression was regulated by other faradic by-products. This study reveals how different electrical stimulation regime alters cellular regenerative responses and the roles of faradic by-products, that might be used as a physical tool to guide and control cell behaviour.
Collapse
Affiliation(s)
- Kasama Srirussamee
- Department of Materials, The University of Manchester, Manchester, UK.,Department of Biomedical Engineering, Faculty of Engineering, King Mongkut's Institute of Technology Ladkrabang (KMITL), Bangkok, Thailand
| | - Ruikang Xue
- Department of Materials, The University of Manchester, Manchester, UK
| | - Sahba Mobini
- Department of Materials, The University of Manchester, Manchester, UK.,Instituto de Micro y Nanotecnología IMN-CNM, The Spanish National Research Council (CSIC), Madrid, Comunidad de Madrid, Spain.,Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Nigel J Cassidy
- Department of Civil Engineering, University of Birmingham, Birmingham, UK
| | - Sarah H Cartmell
- Department of Materials, The University of Manchester, Manchester, UK
| |
Collapse
|
24
|
Rathinam E, Govindarajan S, Rajasekharan S, Declercq H, Elewaut D, De Coster P, Martens L, Leybaert L. The calcium dynamics of human dental pulp stem cells stimulated with tricalcium silicate-based cements determine their differentiation and mineralization outcome. Sci Rep 2021; 11:645. [PMID: 33436827 PMCID: PMC7804324 DOI: 10.1038/s41598-020-80096-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022] Open
Abstract
Calcium (Ca2+) signalling plays an indispensable role in dental pulp and dentin regeneration, but the Ca2+ responses of human dental pulp stem cells (hDPSCs) stimulated with tricalcium silicate-based (TCS-based) dental biomaterials remains largely unexplored. The objective of the present study was to identify and correlate extracellular Ca2+ concentration, intracellular Ca2+ dynamics, pH, cytotoxicity, gene expression and mineralization ability of human dental pulp stem cells (hDPSCs) stimulated with two different TCS-based biomaterials: Biodentine and ProRoot white MTA. The hDPSCs were exposed to the biomaterials, brought in contact with the overlaying medium, with subsequent measurements of extracellular Ca2+ and pH, and intracellular Ca2+ changes. Messenger RNA expression (BGLAP, TGF-β, MMP1 and BMP2), cytotoxicity (MTT and TUNEL) and mineralization potential (Alizarin red and Von Kossa staining) were then evaluated. Biodentine released significantly more Ca2+ in the α-MEM medium than ProRoot WMTA but this had no cytotoxic impact on hDPSCs. The larger Biodentine-linked Ca2+ release resulted in altered intracellular Ca2+ dynamics, which attained a higher maximum amplitude, faster rise time and increased area under the curve of the Ca2+ changes compared to ProRoot WMTA. Experiments with intracellular Ca2+ chelation, demonstrated that the biomaterial-triggered Ca2+ dynamics affected stem cell-related gene expression, cellular differentiation and mineralization potential. In conclusion, biomaterial-specific Ca2+ dynamics in hDPSCs determine differentiation and mineralization outcomes, with increased Ca2+ dynamics enhancing mineralization.
Collapse
Affiliation(s)
- Elanagai Rathinam
- Department of Paediatric Dentistry and Special Care, PAECOMEDIS Research Cluster, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium.
| | - Srinath Govindarajan
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium.,Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium
| | - Sivaprakash Rajasekharan
- Department of Paediatric Dentistry and Special Care, PAECOMEDIS Research Cluster, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium
| | - Heidi Declercq
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium.,Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, 8500, Kortrijk, Belgium
| | - Dirk Elewaut
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium.,Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium
| | - Peter De Coster
- Department of Reconstructive Dentistry and Oral Biology, Dental School, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium
| | - Luc Martens
- Department of Paediatric Dentistry and Special Care, PAECOMEDIS Research Cluster, Ghent University, Ghent University Hospital, 9000, Ghent, Belgium
| | - Luc Leybaert
- Department of Basic And Applied Medical Sciences - Physiology Group, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Gavazzo P, Viti F, Donnelly H, Oliva MAG, Salmeron-Sanchez M, Dalby MJ, Vassalli M. Biophysical phenotyping of mesenchymal stem cells along the osteogenic differentiation pathway. Cell Biol Toxicol 2021; 37:915-933. [PMID: 33420657 DOI: 10.1007/s10565-020-09569-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/30/2020] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells represent an important resource, for bone regenerative medicine and therapeutic applications. This review focuses on new advancements and biophysical tools which exploit different physical and chemical markers of mesenchymal stem cell populations, to finely characterize phenotype changes along their osteogenic differentiation process. Special attention is paid to recently developed label-free methods, which allow monitoring cell populations with minimal invasiveness. Among them, quantitative phase imaging, suitable for single-cell morphometric analysis, and nanoindentation, functional to cellular biomechanics investigation. Moreover, the pool of ion channels expressed in cells during differentiation is discussed, with particular interest for calcium homoeostasis.Altogether, a biophysical perspective of osteogenesis is proposed, offering a valuable tool for the assessment of the cell stage, but also suggesting potential physiological links between apparently independent phenomena.
Collapse
Affiliation(s)
- Paola Gavazzo
- Institute of Biophysics, National Research Council, Genoa, Italy
| | - Federica Viti
- Institute of Biophysics, National Research Council, Genoa, Italy.
| | - Hannah Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Mariana Azevedo Gonzalez Oliva
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| |
Collapse
|
26
|
Ryan CNM, Doulgkeroglou MN, Zeugolis DI. Electric field stimulation for tissue engineering applications. BMC Biomed Eng 2021; 3:1. [PMID: 33397515 PMCID: PMC7784019 DOI: 10.1186/s42490-020-00046-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/06/2020] [Indexed: 01/02/2023] Open
Abstract
Electric fields are involved in numerous physiological processes, including directional embryonic development and wound healing following injury. To study these processes in vitro and/or to harness electric field stimulation as a biophysical environmental cue for organised tissue engineering strategies various electric field stimulation systems have been developed. These systems are overall similar in design and have been shown to influence morphology, orientation, migration and phenotype of several different cell types. This review discusses different electric field stimulation setups and their effect on cell response.
Collapse
Affiliation(s)
- Christina N M Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Ireland Galway & USI, Galway, Ireland.,Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Meletios N Doulgkeroglou
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Ireland Galway & USI, Galway, Ireland.,Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), National University of Ireland Galway & USI, Galway, Ireland. .,Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland. .,Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland.
| |
Collapse
|
27
|
Electrical Stimulation of Adipose-Derived Stem Cells in 3D Nanofibrillar Cellulose Increases Their Osteogenic Potential. Biomolecules 2020; 10:biom10121696. [PMID: 33353222 PMCID: PMC7766661 DOI: 10.3390/biom10121696] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Due to the ageing population, there is a steadily increasing incidence of osteoporosis and osteoporotic fractures. As conventional pharmacological therapy options for osteoporosis are often associated with severe side effects, bone grafts are still considered the clinical gold standard. However, the availability of viable, autologous bone grafts is limited making alternative cell-based strategies a promising therapeutic alternative. Adipose-derived stem cells (ASCs) are a readily available population of mesenchymal stem/stromal cells (MSCs) that can be isolated within minimally invasive surgery. This ease of availability and their ability to undergo osteogenic differentiation makes ASCs promising candidates for cell-based therapies for bone fractures. Recent studies have suggested that both exposure to electrical fields and cultivation in 3D can positively affect osteogenic potential of MSCs. To elucidate the osteoinductive potential of a combination of these biophysical cues on ASCs, cells were embedded within anionic nanofibrillar cellulose (aNFC) hydrogels and exposed to electrical stimulation (ES) for up to 21 days. ES was applied to ASCs in 2D and 3D at a voltage of 0.1 V/cm with a duration of 0.04 ms, and a frequency of 10 Hz for 30 min per day. Exposure of ASCs to ES in 3D resulted in high alkaline phosphatase (ALP) activity and in an increased mineralisation evidenced by Alizarin Red S staining. Moreover, ES in 3D aNFC led to an increased expression of the osteogenic markers osteopontin and osteocalcin and a rearrangement and alignment of the actin cytoskeleton. Taken together, our data suggest that a combination of ES with 3D cell culture can increase the osteogenic potential of ASCs. Thus, exposure of ASCs to these biophysical cues might improve the clinical outcomes of regenerative therapies in treatment of osteoporotic fractures.
Collapse
|
28
|
Lai YS, Chang YH, Chen YY, Xu J, Yu CS, Chang SJ, Chen PS, Tsai SJ, Chiu WT. Ca 2+ -regulated cell migration revealed by optogenetically engineered Ca 2+ oscillations. J Cell Physiol 2020; 236:4681-4693. [PMID: 33244795 PMCID: PMC8048425 DOI: 10.1002/jcp.30190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 01/05/2023]
Abstract
The ability of a single Ca2+ ion to play an important role in cell biology is highlighted by the need for cells to form Ca2+ signals in the dimensions of space, time, and amplitude. Thus, spatial and temporal changes in intracellular Ca2+ concentration are important for determining cell fate. Optogenetic technology has been developed to provide more precise and targeted stimulation of cells. Here, U2OS cells overexpressing Ca2+ translocating channelrhodopsin (CatCh) were used to mediate Ca2+ influx through blue light illumination with various parameters, such as intensity, frequency, duty cycle, and duration. We identified that several Ca2+‐dependent transcription factors and certain kinases can be activated by specific Ca2+ waves. Using a wound‐healing assay, we found that low‐frequency Ca2+ oscillations increased cell migration through the activation of NF‐κB. This study explores the regulation of cell migration by Ca2+ signals. Thus, we can choose optical parameters to modulate Ca2+ waves and achieve activation of specific signaling pathways. This novel methodology can be applied to clarify related cell‐signaling mechanisms in the future.
Collapse
Affiliation(s)
- Yi-Shyun Lai
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Han Chang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yong-Yi Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Jixuan Xu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Sian Yu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Su-Jing Chang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Pai-Sheng Chen
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
29
|
Ayad O, Al Sayed ZR, Sebille S, Magaud C, Chapotte-Baldacci CA, Jayle C, Faivre JF, Gaborit N, Chatelier A, Bois P. In vitro differentiation of W8B2 + human cardiac stem cells: gene expression of ionic channels and spontaneous calcium activity. Cell Mol Biol Lett 2020; 25:50. [PMID: 33292162 PMCID: PMC7646077 DOI: 10.1186/s11658-020-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022] Open
Abstract
Background Human cardiac stem cells expressing the W8B2 marker (W8B2+ CSCs) were recently identified and proposed as a new model of multipotent CSCs capable of differentiating into smooth muscle cells, endothelial cells and immature myocytes. Nevertheless, no characterization of ion channel or calcium activity during the differentiation of these stem cells has been reported. Methods The objectives of this study were thus to analyze (using the TaqMan Low-Density Array technique) the gene profile of W8B2+ CSCs pertaining to the regulation of ion channels, transporters and other players involved in the calcium homeostasis of these cells. We also analyzed spontaneous calcium activity (via the GCaMP calcium probe) during the in vitro differentiation of W8B2+ CSCs into cardiac myocytes. Results Our results show an entirely different electrophysiological genomic profile between W8B2+ CSCs before and after differentiation. Some specific nodal genes, such as Tbx3, HCN, ICaT, L, KV, and NCX, are overexpressed after this differentiation. In addition, we reveal spontaneous calcium activity or a calcium clock whose kinetics change during the differentiation process. A pharmacological study carried out on differentiated W8B2+ CSCs showed that the NCX exchanger and IP3 stores play a fundamental role in the generation of these calcium oscillations. Conclusions Taken together, the present results provide important information on ion channel expression and intrinsic calcium dynamics during the differentiation process of stem cells expressing the W8B2 marker.
Collapse
Affiliation(s)
- Oualid Ayad
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Zeina R Al Sayed
- CNRS, INSERM, l'institut du thorax, Université de Nantes, 44000, Nantes, France
| | - Stéphane Sebille
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Christophe Magaud
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | | | - Christophe Jayle
- CHU of Poitiers chirurgie cardiaque et thoracique, , Poitiers Cedex 09, France
| | - Jean-François Faivre
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Nathalie Gaborit
- CNRS, INSERM, l'institut du thorax, Université de Nantes, 44000, Nantes, France
| | - Aurélien Chatelier
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Patrick Bois
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France.
| |
Collapse
|
30
|
Stem cell plasticity and regenerative potential regulation through Ca 2+-mediated mitochondrial nuclear crosstalk. Mitochondrion 2020; 56:1-14. [PMID: 33059088 DOI: 10.1016/j.mito.2020.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/03/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The multi-lineage differentiation potential is one of the prominent mechanisms through which stem cells can repair damaged tissues. The regenerative potential of stem cells is the manifestation of several changes at the structural and molecular levels in stem cells that are regulated through intricate mitochondrial-nuclear interactions maintained by Ca2+ ion signaling. Despite the exhilarating evidences strengthening the versatile and indispensible role of Ca2+ in regulating mitochondrial-nuclear interactions, the extensive details of signaling mechanisms remains largely unexplored. In this review we have discussed the effect of Ca2+ ion mediated mitochondrial-nuclear interactions participating in stem plasticity and its regenerative potential.
Collapse
|
31
|
De Angelis A, Denzi A, Merla C, Andre FM, Mir LM, Apollonio F, Liberti M. Confocal Microscopy Improves 3D Microdosimetry Applied to Nanoporation Experiments Targeting Endoplasmic Reticulum. Front Bioeng Biotechnol 2020; 8:552261. [PMID: 33072718 PMCID: PMC7537786 DOI: 10.3389/fbioe.2020.552261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
In the last years, microdosimetric numerical models of cells including intracellular compartments have been proposed, aiming to investigate the poration induced by the application of nanosecond pulsed electric fields (nsPEFs). A limitation of such models was the extremely approximate cell and organelle shapes, leading to an incorrect estimation of the electric field or transmembrane potential distribution in the studied domain. In order to obtain a reliable model of in vitro experiments and a one-to-one comparison between experimental and simulated results, here, a realistic model of 12 human mesenchymal stem cells was built starting from their optical microscopy images where different cell compartments were highlighted. The microdosimetric analysis of the cells group was quantified in terms of electric field and transmembrane potentials (TMPs) induced by an externally applied 10-ns trapezoidal pulse with rise and fall times of 2 ns, with amplitudes ranging from 2 to 30 MV/m. The obtained results showed that the plasma and endoplasmic reticulum (ER) membrane of each cell respond in a different way to the same electric field amplitude, depending on differences in shape, size, and position of the single cell with respect to the applied electric field direction. Therefore, also the threshold for an efficient electroporation is highly different from cell to cell. This difference was quantitatively estimated through the cumulative distribution function of the pore density for the plasma and ER membrane of each cell, representing the probability that a certain percentage of membrane has reached a specific value of pore density. By comparing the dose-response curves resulted from the simulations and those from the experimental study of De Menorval et al. (2016), we found a very good matching of results for plasma and ER membrane when 2% of the porated area is considered sufficient for permeabilizing the membrane. This result is worth of noting as it highlights the possibility to effectively predict the behavior of a cell (or of a population of cells) exposed to nsPEFs. Therefore, the microdosimetric realistic model described here could represent a valid tool in setting up more efficient and controlled electroporation protocols.
Collapse
Affiliation(s)
- Annalisa De Angelis
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Agnese Denzi
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy
| | - Caterina Merla
- National Italian Agency for Energy, New Technologies and Sustainable Economic Development - Department of Sustainability (ENEA, SSPT) - Division of Health Protection Technologies, Rome, Italy
| | - Frank M Andre
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Metabolic and Systemic Aspects of Oncogenesis, Villejuif, France
| | - Lluis M Mir
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Metabolic and Systemic Aspects of Oncogenesis, Villejuif, France
| | - Francesca Apollonio
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Micaela Liberti
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| |
Collapse
|
32
|
Rahmani A, Naderi M, Barati G, Arefian E, Jedari B, Nadri S. The potency of hsa-miR-9-1 overexpression in photoreceptor differentiation of conjunctiva mesenchymal stem cells on a 3D nanofibrous scaffold. Biochem Biophys Res Commun 2020; 529:526-532. [PMID: 32736669 DOI: 10.1016/j.bbrc.2020.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/03/2020] [Indexed: 12/31/2022]
Abstract
MiRNAs are small non-coding RNAs that are ordinarily involved in modulating mRNAs and stem cell differentiation. 3D nanofibrous scaffolds have an important role in the differentiation of stem cells due to their similarity to the extracellular matrix (ECM). In the present study, we tried to introduce a new approach to guiding the differentiation of conjunctiva mesenchymal stem cells (CJMSCs) into photoreceptor-like cells by hsa-miR-9-1 delivery on both 2D and 3D substrates. First, the CJMSCs were transduced by a lentiviral vector carrying miR-9 (pCDH + hsa-miR-9-1) and then cell transduction efficacy verified by using fluorescent microscopy, flow cytometry, and qPCR analyses. Silk Fibroin-poly-L-lactic acid (SF-PLLA) scaffold was fabricated by the electrospinning technique while the scaffold characteristics including morphology, chemical properties, and biocompatibility were evaluated by SEM, FTIR, and MTT assays, respectively. Then, the miR-9-CJMSCs were seeded on both TCPS and the scaffold; photoreceptor gene and protein expressions were evaluated by RT-qPCR and immunostaining after 14 and 21 days of transduction. More than 80% of CJMSCs were transduced and miR-9 expression was significantly higher in miR-9-CJMSCs compared with empty vector (EV)-CJMSCs. SEM and FTIR confirmed the fabrication of the SF/PLLA hybrid structure. RT-qPCR and immunostaining analyses showed that the specific photoreceptor genes and proteins were expressed in miR-9 transduced CJMSCs. Mir-9 induced CJMSCs into photoreceptor-like cells in a time-dependent manneron on both TCPS and nanofibrous scaffold.We have proved that hsa-miR-9-1 has the potency to guide the photoreceptor differentiation of mesenchymal stem cells and promote retinal regeneration.
Collapse
Affiliation(s)
- Ali Rahmani
- Department of Medical Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahmood Naderi
- Cell-Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Barati
- Department of Medical Biotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Iran
| | - Behrouz Jedari
- Department of Medical Biotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Department of Medical Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
33
|
Venkateswarlu K, Suman G, Dhyani V, Swain S, Giri L, Samavedi S. Three‐dimensional imaging and quantification of real‐time cytosolic calcium oscillations in microglial cells cultured on electrospun matrices using laser scanning confocal microscopy. Biotechnol Bioeng 2020; 117:3108-3123. [DOI: 10.1002/bit.27465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 05/24/2020] [Accepted: 06/16/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Kojja Venkateswarlu
- Department of Chemical Engineering Indian Institute of Technology Hyderabad Sangareddy Telangana India
| | - Gare Suman
- Department of Chemical Engineering Indian Institute of Technology Hyderabad Sangareddy Telangana India
| | - Vaibhav Dhyani
- Department of Chemical Engineering Indian Institute of Technology Hyderabad Sangareddy Telangana India
| | - Sarpras Swain
- Department of Chemical Engineering Indian Institute of Technology Hyderabad Sangareddy Telangana India
| | - Lopamudra Giri
- Department of Chemical Engineering Indian Institute of Technology Hyderabad Sangareddy Telangana India
| | - Satyavrata Samavedi
- Department of Chemical Engineering Indian Institute of Technology Hyderabad Sangareddy Telangana India
| |
Collapse
|
34
|
Leppik L, Oliveira KMC, Bhavsar MB, Barker JH. Electrical stimulation in bone tissue engineering treatments. Eur J Trauma Emerg Surg 2020; 46:231-244. [PMID: 32078704 PMCID: PMC7113220 DOI: 10.1007/s00068-020-01324-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Electrical stimulation (EStim) has been shown to promote bone healing and regeneration both in animal experiments and clinical treatments. Therefore, incorporating EStim into promising new bone tissue engineering (BTE) therapies is a logical next step. The goal of current BTE research is to develop combinations of cells, scaffolds, and chemical and physical stimuli that optimize treatment outcomes. Recent studies demonstrating EStim's positive osteogenic effects at the cellular and molecular level provide intriguing clues to the underlying mechanisms by which it promotes bone healing. In this review, we discuss results of recent in vitro and in vivo research focused on using EStim to promote bone healing and regeneration and consider possible strategies for its application to improve outcomes in BTE treatments. Technical aspects of exposing cells and tissues to EStim in in vitro and in vivo model systems are also discussed.
Collapse
Affiliation(s)
- Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany.
| | - Karla Mychellyne Costa Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - Mit Balvantray Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - John Howard Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
35
|
Investigation of The Cellular Response to Bone Fractures: Evidence for Flexoelectricity. Sci Rep 2020; 10:254. [PMID: 31937885 PMCID: PMC6959267 DOI: 10.1038/s41598-019-57121-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/20/2019] [Indexed: 01/08/2023] Open
Abstract
The recent discovery of bone flexoelectricity (strain-gradient-induced electrical polarization) suggests that flexoelectricity could have physiological effects in bones, and specifically near bone fractures, where flexoelectricity is theoretically highest. Here, we report a cytological study of the interaction between crack stress and bone cells. We have cultured MC3T3-E1 mouse osteoblastic cells in biomimetic microcracked hydroxyapatite substrates, differentiated into osteocytes and applied a strain gradient to the samples. The results show a strong apoptotic cellular response, whereby mechanical stimulation causes those cells near the crack to die, as indicated by live-dead and caspase staining. In addition, analysis two weeks post-stimulation shows increased cell attachment and mineralization around microcracks and a higher expression of osteocalcin –an osteogenic protein known to be promoted by physical exercise. The results are consistent with flexoelectricity playing at least two different roles in bone remodelling: apoptotic trigger of the repair protocol, and electro-stimulant of the bone-building activity of osteoblasts.
Collapse
|
36
|
Abstract
Stem cells can be conceptualized as computational processors capable of sensing, processing, and converting environmental information (input) to yield a specific differentiation pathway (output). In this study, we employ a temperature-controlled polymer sheet actuator to interpret and transfer information, controlled by the material’s programming, to mesenchymal stem cells. The cell’s interpretation of mechanical, thermal, and biochemical signaling is shown to be dependent on the actuator’s activity, utilized to accelerate differentiation toward bone cells, further elucidating the role of microenvironmental parameters on mammalian cells. Our method provides a unique approach to processing two discrete stimuli into one biochemical signal, calcium ions, providing a basis for the logical control of the flow of biological signals and the design of cellular functions. Stem cells are capable of sensing and processing environmental inputs, converting this information to output a specific cell lineage through signaling cascades. Despite the combinatorial nature of mechanical, thermal, and biochemical signals, these stimuli have typically been decoupled and applied independently, requiring continuous regulation by controlling units. We employ a programmable polymer actuator sheet to autonomously synchronize thermal and mechanical signals applied to mesenchymal stem cells (MSCs). Using a grid on its underside, the shape change of polymer sheet, as well as cell morphology, calcium (Ca2+) influx, and focal adhesion assembly, could be visualized and quantified. This paper gives compelling evidence that the temperature sensing and mechanosensing of MSCs are interconnected via intracellular Ca2+. Up-regulated Ca2+ levels lead to a remarkable alteration of histone H3K9 acetylation and activation of osteogenic related genes. The interplay of physical, thermal, and biochemical signaling was utilized to accelerate the cell differentiation toward osteogenic lineage. The approach of programmable bioinstructivity provides a fundamental principle for functional biomaterials exhibiting multifaceted stimuli on differentiation programs. Technological impact is expected in the tissue engineering of periosteum for treating bone defects.
Collapse
|
37
|
Ross CL, Zhou Y, McCall CE, Soker S, Criswell TL. The Use of Pulsed Electromagnetic Field to Modulate Inflammation and Improve Tissue Regeneration: A Review. Bioelectricity 2019; 1:247-259. [PMID: 34471827 PMCID: PMC8370292 DOI: 10.1089/bioe.2019.0026] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pulsed electromagnetic field (PEMF) is emerging as innovative treatment for regulation of inflammation, which could have significant effects on tissue regeneration. PEMF modulates inflammatory processes through the regulation of pro- and anti-inflammatory cytokine secretion during different stages of inflammatory response. Consistent outcomes in studies involving animal and human tissue have shown promise for the use of PEMF as an alternative or complementary treatment to pharmaceutical therapies. Thus, PEMF treatment could provide a novel nonpharmaceutical means of modulating inflammation in injured tissues resulting in enhanced functional recovery. This review examines the effect of PEMF on immunomodulatory cells (e.g., mesenchymal stem/stromal cells [MSCs] and macrophages [MΦ]) to better understand the potential for PEMF therapy to modulate inflammatory signaling pathways and improve tissue regeneration. This review cites published data that support the use of PEMF to improve tissue regeneration. Our studies included herein confirm anti-inflammatory effects of PEMF on MSCs and MΦ.
Collapse
Affiliation(s)
- Christina L. Ross
- Center for Integrative Medicine, Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Yu Zhou
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Charles E. McCall
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Shay Soker
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tracy L. Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
38
|
Fu J, Liu X, Tan L, Cui Z, Zheng Y, Liang Y, Li Z, Zhu S, Yeung KWK, Feng X, Wang X, Wu S. Photoelectric-Responsive Extracellular Matrix for Bone Engineering. ACS NANO 2019; 13:13581-13594. [PMID: 31697055 DOI: 10.1021/acsnano.9b08115] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Using noninvasive stimulation of cells to control cell fate and improve bone regeneration by optical stimulation can achieve the aim of precisely orchestrating biological activities. In this study, we create a fast and repeatable photoelectric-responsive microenvironment around an implant using a bismuth sulfide/hydroxyapatite (BS/HAp) film. The unexpected increase of photocurrent on the BS/HAp film under near-infrared (NIR) light is mainly due to the depletion of holes through PO43- from HAp and interfacial charge transfer by HAp compared with BS. The electrons activate the Na+ channel of mesenchymal stem cells (MSCs) and change the cell adhesion in the intermediate environment. The behavior of MSCs is tuned by changing the photoelectronic microenvironment. RNA sequencing reveals that when photoelectrons transfer to the cell membrane, sodium ions flux and the membrane potential depolarizes to change the cell shape. Meanwhile, calcium ions fluxed and FDE1 was upregulated. Furthermore, the TCF/LEF in the cell nucleus began transcription to regulate the downstream genes involved in osteogenic differentiation, which is performed through the Wnt/Ca2+ signaling pathway. This research has created a biological therapeutic strategy, which can achieve in vitro remotely, precisely, and noninvasively controlling cell differentiation behaviors by tuning the in vivo photoelectric microenvironment using NIR light.
Collapse
Affiliation(s)
- Jieni Fu
- Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , People's Republic of China
| | - Xiangmei Liu
- Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , People's Republic of China
| | - Lei Tan
- Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , People's Republic of China
| | - Zhenduo Cui
- School of Materials Science & Engineering, Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China , Tianjin University , Tianjin 300072 , People's Republic of China
| | - Yufeng Zheng
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering , Peking University , Beijing 100871 , People's Republic of China
| | - Yanqin Liang
- School of Materials Science & Engineering, Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China , Tianjin University , Tianjin 300072 , People's Republic of China
| | - Zhaoyang Li
- School of Materials Science & Engineering, Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China , Tianjin University , Tianjin 300072 , People's Republic of China
| | - Shengli Zhu
- School of Materials Science & Engineering, Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China , Tianjin University , Tianjin 300072 , People's Republic of China
| | - Kelvin Wai Kwok Yeung
- Department of Orthopaedics & Traumatology, Li KaShing Faculty of Medicine , The University of Hong Kong , Pokfulam , Hong Kong 999077 , People's Republic of China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , People's Republic of China
| | - Xianbao Wang
- Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , People's Republic of China
| | - Shuilin Wu
- Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , People's Republic of China
- School of Materials Science & Engineering, Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China , Tianjin University , Tianjin 300072 , People's Republic of China
| |
Collapse
|
39
|
Optically Transparent Anionic Nanofibrillar Cellulose Is Cytocompatible with Human Adipose Tissue-Derived Stem Cells and Allows Simple Imaging in 3D. Stem Cells Int 2019; 2019:3106929. [PMID: 31687032 PMCID: PMC6800951 DOI: 10.1155/2019/3106929] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
The anti-inflammatory and immunomodulatory properties of human mesenchymal stromal cells (MSCs) are a focus within regenerative medicine. However, 2D cultivation of MSCs for extended periods results in abnormal cell polarity, chromosomal changes, reduction in viability, and altered differentiation potential. As an alternative, various 3D hydrogels have been developed which mimic the endogenous niche of MSCs. Nevertheless, imaging cells embedded within 3D hydrogels often suffers from low signal-to-noise ratios which can be at least partly attributed to the high light absorbance and light scattering of the hydrogels in the visible light spectrum. In this study, human adipose tissue-derived MSCs (ADSCs) are cultivated within an anionic nanofibrillar cellulose (aNFC) hydrogel. It is demonstrated that aNFC forms nanofibres arranged as a porous network with low light absorbance in the visible spectrum. Moreover, it is shown that aNFC is cytocompatible, allowing for MSC proliferation, maintaining cell viability and multilineage differentiation potential. Finally, aNFC is compatible with scanning electron microscopy (SEM) and light microscopy including the application of conventional dyes, fluorescent probes, indirect immunocytochemistry, and calcium imaging. Overall, the results indicate that aNFC represents a promising 3D material for the expansion of MSCs whilst allowing detailed examination of cell morphology and cellular behaviour.
Collapse
|
40
|
Srirussamee K, Mobini S, Cassidy NJ, Cartmell SH. Direct electrical stimulation enhances osteogenesis by inducing Bmp2 and Spp1 expressions from macrophages and preosteoblasts. Biotechnol Bioeng 2019; 116:3421-3432. [PMID: 31429922 PMCID: PMC6899728 DOI: 10.1002/bit.27142] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/03/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022]
Abstract
The capability of electrical stimulation (ES) in promoting bone regeneration has already been addressed in clinical studies. However, its mechanism is still being investigated and discussed. This study aims to investigate the responses of macrophages (J774A.1) and preosteoblasts (MC3T3-E1) to ES and the faradic by-products from ES. It is found that pH of the culture media was not significantly changed, whereas the average hydrogen peroxide concentration was increased by 3.6 and 5.4 µM after 1 and 2 hr of ES, respectively. The upregulation of Bmp2 and Spp1 messenger RNAs was observed after 3 days of stimulation, which is consistent among two cell types. It is also found that Spp1 expression of macrophages was partially enhanced by faradic by-products. Osteogenic differentiation of preosteoblasts was not observed during the early stage of ES as the level of Runx2 expression remains unchanged. However, cell proliferation was impaired by the excessive current density from the electrodes, and also faradic by-products in the case of macrophages. This study shows that macrophages could respond to ES and potentially contribute to the bone formation alongside preosteoblasts. The upregulation of Bmp2 and Spp1 expressions induced by ES could be one of the mechanisms behind the electrically stimulated osteogenesis.
Collapse
Affiliation(s)
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología IMN-CNM, The Spanish National Research Council (CSIC), Madrid, Spain.,Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Nigel J Cassidy
- Department of Civil Engineering, University of Birmingham, Birmingham, UK
| | - Sarah H Cartmell
- Department of Materials, The University of Manchester, Manchester, UK
| |
Collapse
|
41
|
Cruciani S, Santaniello S, Montella A, Ventura C, Maioli M. Orchestrating stem cell fate: Novel tools for regenerative medicine. World J Stem Cells 2019; 11:464-475. [PMID: 31523367 PMCID: PMC6716083 DOI: 10.4252/wjsc.v11.i8.464] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells are undifferentiated cells able to acquire different phenotypes under specific stimuli. In vitro manipulation of these cells is focused on understanding stem cell behavior, proliferation and pluripotency. Latest advances in the field of stem cells concern epigenetics and its role in maintaining self-renewal and differentiation capabilities. Chemical and physical stimuli can modulate cell commitment, acting on gene expression of Oct-4, Sox-2 and Nanog, the main stemness markers, and tissue-lineage specific genes. This activation or repression is related to the activity of chromatin-remodeling factors and epigenetic regulators, new targets of many cell therapies. The aim of this review is to afford a view of the current state of in vitro and in vivo stem cell applications, highlighting the strategies used to influence stem cell commitment for current and future cell therapies. Identifying the molecular mechanisms controlling stem cell fate could open up novel strategies for tissue repairing processes and other clinical applications.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, Bologna 40129, Italy
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, Bologna 40129, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
- Operative Unit of Clinical Genetics and Developmental Biology, Sassari 07100, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, Bologna 40129, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems – Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, Bologna 40129, Italy
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari 09042, Italy
- Center for Developmental Biology and Reprogramming-CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy.
| |
Collapse
|
42
|
Sohn JO, Seong SY, Kim HJ, Jo YM, Lee KH, Chung MK, Song HJ, Park KS, Lim JM. Alterations in intracellular Ca 2+ levels in human endometrial stromal cells after decidualization. Biochem Biophys Res Commun 2019; 515:318-324. [PMID: 31153638 DOI: 10.1016/j.bbrc.2019.05.153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 01/17/2023]
Abstract
Calcium (Ca2+) is an important element for many physiological functions of the uterus, including embryo implantation. Here, we investigated the possible involvement of altered intracellular Ca2+ levels in decidualization in human endometrial stromal cells (hEMSCs). hEMSCs showed high levels of mesenchymal stem cell marker expression (CD73, CD90, and CD105) and did not express markers of hematopoietic progenitor cells (CD31, CD34, CD45, and HLA-DR). Decidualization is a process of ovarian steroid-induced endometrial stromal cell proliferation and differentiation. Several types of ion channels, which are regulated by the ovarian hormones progesterone and estradiol, as well as growth factors, are important for endometrial receptivity and embryo implantation. The combined application of progesterone (1 μM medroxyprogesterone acetate) and cyclic AMP (0.5 mM) for 6 days not only elevated inositol 1,4,5-triphosphate receptor (IP3R)-mediated Ca2+ release and IP3R expression, it also promoted ORAI and STIM expression as well as cyclopiazonic acid-induced Ca2+ release. Finally, intracellular Ca2+ levels and ion channel gene expression influenced hEMSC proliferation. These results suggest that cytosolic Ca2+ dynamics, mediated by specific ion channels, serve as an important step in the decidualization of hEMSCs.
Collapse
Affiliation(s)
- Jie Ohn Sohn
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 151-921, South Korea; Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Seung Yong Seong
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, 25159, South Korea
| | - Hyun Jin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Yoon Mi Jo
- Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Kyoung Hoon Lee
- Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Mi Kyung Chung
- Seoul Rachel Fertility Center, Seoul, 04146, South Korea
| | - Hyun Jin Song
- Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Kyoung Sun Park
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, 25159, South Korea.
| | - Jeong Mook Lim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 151-921, South Korea; Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, South Korea.
| |
Collapse
|
43
|
Vickers L, Thorpe AA, Snuggs J, Sammon C, Le Maitre CL. Mesenchymal stem cell therapies for intervertebral disc degeneration: Consideration of the degenerate niche. JOR Spine 2019; 2:e1055. [PMID: 31463465 PMCID: PMC6686825 DOI: 10.1002/jsp2.1055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
We have previously reported a synthetic Laponite crosslinked poly N-isopropylacrylamide-co-N, N'-dimethylacrylamide (NPgel) hydrogel, which induces nucleus pulposus (NP) cell differentiation of human mesenchymal stem cells (hMSCs) without the need for additional growth factors. Furthermore NP gel supports integration following injection into the disc and restores mechanical function to the disc. However, translation of this treatment strategy into clinical application is dependent on the survival and differentiation of hMSC to the correct cell phenotype within the degenerate intervertebral disc (IVD). Here, we investigated the viability and differentiation of hMSCs within NP gel within a catabolic microenvironment. hMSCs were encapsulated in NPgel and cultured for 4 weeks under hypoxia (5% O2) with ± calcium, interleukin-1β (IL-1β), and tumor necrosis factor alpha (TNFα) either individually or in combination to mimic the degenerate environment. Cell viability and cellular phenotype were investigated. Stem cell viability was maintained within hydrogel systems for the 4 weeks investigated under all degenerate conditions. NP matrix markers: Agg and Col II and NP phenotypic markers: HIF-1α, FOXF1, and PAX1 were expressed within the NPgel cultures and expression was not affected by culture within degenerate conditions. Alizarin red staining demonstrated increased calcium deposition under cultures containing CaCl2 indicating calcification of the matrix. Interestingly matrix metalloproteinases (MMPs), ADAMTS 4, and Col I expression by hMSCs cultured in NPgel was upregulated by calcium but not by proinflammatory cytokines IL-1β and TNFα. Importantly IL-1β and TNFα, regarded as key contributors to disc degeneration, were not shown to affect the NP cell differentiation of mesenchymal stem cells (MSCs) in the NPgel. In agreement with our previous findings, NPgel alone was sufficient to induce NP cell differentiation of MSCs, with expression of both aggrecan and collagen type II, under both standard and degenerate culture conditions; thus could provide a therapeutic option for the repair of the NP during IVD degeneration.
Collapse
Affiliation(s)
- Louise Vickers
- Biomolecular Sciences Research CentreSheffield Hallam UniversitySheffieldUK
| | - Abbey A. Thorpe
- Biomolecular Sciences Research CentreSheffield Hallam UniversitySheffieldUK
| | - Joseph Snuggs
- Biomolecular Sciences Research CentreSheffield Hallam UniversitySheffieldUK
| | - Christopher Sammon
- Materials and Engineering Research InstituteSheffield Hallam UniversitySheffieldUK
| | | |
Collapse
|
44
|
Zheng M, Kim DY, Sung JH. Ion channels and transporters in adipose-derived stem cells. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-018-00413-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
45
|
Li Z, Liu T, Gilmore A, Gómez NM, Mitchell CH, Li YP, Oursler MJ, Yang S. Regulator of G Protein Signaling Protein 12 (Rgs12) Controls Mouse Osteoblast Differentiation via Calcium Channel/Oscillation and Gαi-ERK Signaling. J Bone Miner Res 2019; 34:752-764. [PMID: 30489658 PMCID: PMC7675783 DOI: 10.1002/jbmr.3645] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/13/2018] [Accepted: 11/17/2018] [Indexed: 12/11/2022]
Abstract
Bone homeostasis intimately relies on the balance between osteoblasts (OBs) and osteoclasts (OCs). Our previous studies have revealed that regulator of G protein signaling protein 12 (Rgs12), the largest protein in the Rgs super family, is essential for osteoclastogenesis from hematopoietic cells and OC precursors. However, how Rgs12 regulates OB differentiation and function is still unknown. To understand that, we generated an OB-targeted Rgs12 conditional knockout (CKO) mice model by crossing Rgs12fl/fl mice with Osterix (Osx)-Cre transgenic mice. We found that Rgs12 was highly expressed in both OB precursor cells (OPCs) and OBs of wild-type (WT) mice, and gradually increased during OB differentiation, whereas Rgs12-CKO mice (OsxCre/+ ; Rgs12fl/fl ) exhibited a dramatic decrease in both trabecular and cortical bone mass, with reduced numbers of OBs and increased apoptotic cell population. Loss of Rgs12 in OPCs in vitro significantly inhibited OB differentiation and the expression of OB marker genes, resulting in suppression of OB maturation and mineralization. Further mechanism study showed that deletion of Rgs12 in OPCs significantly inhibited guanosine triphosphatase (GTPase) activity and cyclic adenosine monophosphate (cAMP) level, and impaired Calcium (Ca2+ ) oscillations via restraints of major Ca2+ entry sources (extracellular Ca2+ influx and intracellular Ca2+ release from endoplasmic reticulum), partially contributed by the blockage of L-type Ca2+ channel mediated Ca2+ influx. Downstream mediator extracellular signal-related protein kinase (ERK) was found inactive in OBs of OsxCre/+ ; Rgs12fl/fl mice and in OPCs after Rgs12 deletion, whereas application of pertussis toxin (PTX) or overexpression of Rgs12 could rescue the defective OB differentiation via restoration of ERK phosphorylation. Our findings reveal that Rgs12 is an important regulator during osteogenesis and highlight Rgs12 as a potential therapeutic target for bone disorders. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ziqing Li
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Tongjun Liu
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
- Department of Implantology, Shandong Provincial Key Laboratory of Oral Biomedicine, School of Stomatology, Shandong University
- Department of Stomatology, the Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong province 250000, China
| | - Alyssa Gilmore
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
| | - Néstor Más Gómez
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Department of Physiology, School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Yi-ping Li
- Department of Pathology, University of Alabama in Birmingham, Birmingham, AL 35294, USA
| | - Merry J Oursler
- Department of Medicine, Endocrine Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuying Yang
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- The Penn Center for Musculoskeletal Disorders, University of Pennsylvania Philadelphia, PA 19104, USA
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
| |
Collapse
|
46
|
Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Han HJ. Role of HIF1 α Regulatory Factors in Stem Cells. Int J Stem Cells 2019; 12:8-20. [PMID: 30836734 PMCID: PMC6457711 DOI: 10.15283/ijsc18109] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF1) is a master transcription factor that induces the transcription of genes involved in the metabolism and behavior of stem cells. HIF1-mediated adaptation to hypoxia is required to maintain the pluripotency and survival of stem cells under hypoxic conditions. HIF1 activity is well known to be tightly controlled by the alpha subunit of HIF1 (HIF1α). Understanding the regulatory mechanisms that control HIF1 activity in stem cells will provide novel insights into stem cell biology under hypoxia. Recent research has unraveled the mechanistic details of HIF1α regulating processes, suggesting new strategies for regulating stem cells. This review summarizes recent experimental studies on the role of several regulatory factors (including calcium, 2-oxoglutarate-dependent dioxygenase, microtubule network, importin, and coactivators) in regulating HIF1α activity in stem cells.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| |
Collapse
|
47
|
TRPV4-mediated calcium signaling in mesenchymal stem cells regulates aligned collagen matrix formation and vinculin tension. Proc Natl Acad Sci U S A 2019; 116:1992-1997. [PMID: 30674675 DOI: 10.1073/pnas.1811095116] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Microarchitectural cues drive aligned fibrillar collagen deposition in vivo and in biomaterial scaffolds, but the cell-signaling events that underlie this process are not well understood. Utilizing a multicellular patterning model system that allows for observation of intracellular signaling events during collagen matrix assembly, we investigated the role of calcium (Ca2+) signaling in human mesenchymal stem cells (MSCs) during this process. We observed spontaneous Ca2+ oscillations in MSCs during fibrillar collagen assembly, and hypothesized that the transient receptor potential vanilloid 4 (TRPV4) ion channel, a mechanosensitive Ca2+-permeable channel, may regulate this signaling. Inhibition of TRPV4 nearly abolished Ca2+ signaling at initial stages of collagen matrix assembly, while at later times had reduced but significant effects. Importantly, blocking TRPV4 activity dramatically reduced aligned collagen fibril assembly; conversely, activating TRPV4 accelerated aligned collagen formation. TRPV4-dependent Ca2+ oscillations were found to be independent of pattern shape or subpattern cell location, suggesting this signaling mechanism is necessary for aligned collagen formation but not sufficient in the absence of physical (microarchitectural) cues that force multicellular alignment. As cell-generated mechanical forces are known to be critical to the matrix assembly process, we examined the role of TRPV4-mediated Ca2+ signaling in force generated across the load-bearing focal adhesion protein vinculin within MSCs using an FRET-based tension sensor. Inhibiting TRPV4 decreased tensile force across vinculin, whereas TRPV4 activation caused a dynamic unloading and reloading of vinculin. Together, these findings suggest TRPV4 activity regulates forces at cell-matrix adhesions and is critical to aligned collagen matrix assembly by MSCs.
Collapse
|
48
|
Hou J, Luo T, Chen S, Lin S, Yang MM, Li G, Sun D. Calcium Spike Patterns Reveal Linkage of Electrical Stimulus and MSC Osteogenic Differentiation. IEEE Trans Nanobioscience 2019; 18:3-9. [PMID: 30442614 DOI: 10.1109/tnb.2018.2881004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) are easily obtained multipotent cells that are widely applied in regenerative medicine. Electrical stimulation (ES) has a promoting effect on bone healing and osteogenic differentiation of MSCs. Direct and alternating currents (AC) are extensively used to promote the osteogenic differentiation of MSCs in vivo and in vitro. However, information on conducting effective differentiation remains scarce. In this paper, we propose a method to optimize ES parameters based on calcium spike patterns of MSCs. Calcium spike frequency decreases as the osteogenic differentiation of MSC progresses. Furthermore, we tested various ES parameters through the real-time monitoring of calcium spike patterns. We efficiently initiated the process of osteogenic differentiation in MSCs by using the optimal parameters of AC, including voltage, signal shapes, frequency, and duty time. This method provides a new approach to optimize osteogenic differentiation and is potentially useful in clinical treatment such as of bone fractures.
Collapse
|
49
|
Huang X, Das R, Patel A, Nguyen TD. Physical Stimulations for Bone and Cartilage Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:216-237. [PMID: 30740512 PMCID: PMC6366645 DOI: 10.1007/s40883-018-0064-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022]
Abstract
A wide range of techniques and methods are actively invented by clinicians and scientists who are dedicated to the field of musculoskeletal tissue regeneration. Biological, chemical, and physiological factors, which play key roles in musculoskeletal tissue development, have been extensively explored. However, physical stimulation is increasingly showing extreme importance in the processes of osteogenic and chondrogenic differentiation, proliferation and maturation through defined dose parameters including mode, frequency, magnitude, and duration of stimuli. Studies have shown manipulation of physical microenvironment is an indispensable strategy for the repair and regeneration of bone and cartilage, and biophysical cues could profoundly promote their regeneration. In this article, we review recent literature on utilization of physical stimulation, such as mechanical forces (cyclic strain, fluid shear stress, etc.), electrical and magnetic fields, ultrasound, shock waves, substrate stimuli, etc., to promote the repair and regeneration of bone and cartilage tissue. Emphasis is placed on the mechanism of cellular response and the potential clinical usage of these stimulations for bone and cartilage regeneration.
Collapse
|
50
|
Sato M, Asano T, Hosomichi J, Ono T, Nakata T. Optogenetic manipulation of intracellular calcium by BACCS promotes differentiation of MC3T3-E1 cells. Biochem Biophys Res Commun 2018; 506:716-722. [PMID: 30376992 DOI: 10.1016/j.bbrc.2018.10.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 10/28/2022]
Abstract
Bone remodeling is maintained through the balance between bone formation by osteoblasts and bone resorption by osteoclasts. Previous studies suggested that intracellular Ca2+ signaling plays an important role in the differentiation of osteoblasts; however, the molecular mechanism of Ca2+ signaling in the differentiation of osteoblasts remains unclear. To elucidate the effect of Ca2+ signaling in osteoblasts, we employed an optogenetic tool, blue light-activated Ca2+ channel switch (BACCS). BACCS was used to spatiotemporally control intracellular Ca2+ with blue light stimulation. MC3T3-E1 cells, which have been used as a model of differentiation from preosteoblast to osteoblast, were promoted to differentiate by BACCS expression and rhythmical blue light stimulation. The results indicated that intracellular Ca2+ change from the outside of the cells can regulate signaling for differentiation of MC3T3-E1 cells. Our findings provide evidence that Ca2+ could cause osteoblast differentiation.
Collapse
Affiliation(s)
- Moe Sato
- Department of Orthodontic Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; The Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Toshifumi Asano
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; The Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Takao Nakata
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan; The Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.
| |
Collapse
|