1
|
Shelest A, Alaburda A, Vaiciuleviciute R, Uzieliene I, Bialaglovyte P, Bernotiene E. The Effect of TGF-β3 and IL-1β on L-Type Voltage-Operated Calcium Channels and Calcium Ion Homeostasis in Osteoarthritic Chondrocytes and Human Bone Marrow-Derived Mesenchymal Stem Cells During Chondrogenesis. Pharmaceutics 2025; 17:343. [PMID: 40143007 PMCID: PMC11945166 DOI: 10.3390/pharmaceutics17030343] [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: 01/09/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Transforming growth factor-β (TGF-β) and interleukin 1β (IL-1β) are key regulators of the chondrogenic differentiation, physiology and pathology of cartilage tissue, with TGF-β promoting chondrogenesis and matrix formation, while IL-1β exerts catabolic effects, inhibiting chondrogenesis and contributing to cartilage degradation. Both cytokines alter the intracellular calcium ion (iCa2+) levels; however, the exact pathways are not known. Objectives: This study aimed to evaluate the impact of TGF-β3 and IL-1β on calcium homeostasis in human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and chondrocytes during chondrogenesis. Results: TGF-β3 increased iCa2+ levels in both hBM-MSCs and chondrocytes. Furthermore, TGF-β3 increased the functional activity of L-type voltage-operated calcium channels (L-VOCCs) in hBM-MSCs but not in chondrocytes. TGF-β3 and IL-1β reduced L-VOCCs subunit CaV1.2 (CACNA1C) gene expression in chondrocytes. In hBM-MSCs, TGF-β3 and IL-1β increased SERCA pump (ATP2A2) gene expression, while in chondrocytes, this effect was observed only with TGF-β3. Conclusions: TGF-β3 increases iCa2+ both in osteoarthritic chondrocytes and hBM-MSCs during chondrogenesis. In hBM-MSCs, TGF-β3-mediated elevation in iCa2+ is related to the increased functional activity of L-VOCCs. IL-1β does not change iCa2+ in osteoarthritic chondrocytes and hBM-MSCs; however, it initiates the mechanisms leading to further downregulation of iCa2+ in both types of cells. The differential and cell-specific roles of TGF-β3 and IL-1β in the calcium homeostasis of osteoarthritic chondrocytes and hBM-MSCs during chondrogenesis may provide a new insight into future strategies for cartilage repair and osteoarthritis treatment.
Collapse
Affiliation(s)
- Anastasiia Shelest
- Institute of Biosciences, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Aidas Alaburda
- Institute of Biosciences, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (I.U.); (P.B.); (E.B.)
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (I.U.); (P.B.); (E.B.)
| | - Paulina Bialaglovyte
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (I.U.); (P.B.); (E.B.)
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (I.U.); (P.B.); (E.B.)
- VilniusTech Faculty of Fundamental Sciences, Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, LT-10223 Vilnius, Lithuania
| |
Collapse
|
2
|
Innamorati G, Sanchez-Petidier M, Bergafora G, Codazzi C, Palma V, Camera F, Merla C, André FM, Pedraza M, Moreno Manzano V, Caramazza L, Colella M, Marracino P, Balucani M, Apollonio F, Liberti M, Consales C. Characterization of Mesenchymal and Neural Stem Cells Response to Bipolar Microsecond Electric Pulses Stimulation. Int J Mol Sci 2024; 26:147. [PMID: 39796006 PMCID: PMC11720446 DOI: 10.3390/ijms26010147] [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: 10/14/2024] [Revised: 12/12/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
In the tissue regeneration field, stem cell transplantation represents a promising therapeutic strategy. To favor their implantation, proliferation and differentiation need to be controlled. Several studies have demonstrated that stem cell fate can be controlled by applying continuous electric field stimulation. This study aims to characterize the effect of a specific microsecond electric pulse stimulation (bipolar pulses of 100 µs + 100 µs, delivered for 30 min at an intensity of 250 V/cm) to induce an increase in cell proliferation on mesenchymal stem cells (MSCs) and induced neural stem cells (iNSCs). The effect was evaluated in terms of (i) cell counting, (ii) cell cycle, (iii) gene expression, and (iv) apoptosis. The results show that 24 h after the stimulation, cell proliferation, cell cycle, and apoptosis are not affected, but variation in the expression of specific genes involved in these processes is observed. These results led us to investigate cell proliferation until 72 h from the stimulation, observing an increase in the iNSCs number at this time point. The main outcome of this study is that the microsecond electric pulses can modulate stem cell proliferation.
Collapse
Affiliation(s)
- Giorgia Innamorati
- PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| | - Marina Sanchez-Petidier
- Neural Circuits and Behaviour Laboratory, Fundación Hospital Nacional de Parapléjicos, 45004 Toledo, Spain;
- Metabolic and Systemic Aspects of the Oncogenesis (METSY), CNRS, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France;
| | - Giulia Bergafora
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| | - Camilla Codazzi
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| | - Valentina Palma
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| | - Francesca Camera
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| | - Caterina Merla
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| | - Franck M. André
- Metabolic and Systemic Aspects of the Oncogenesis (METSY), CNRS, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France;
| | - Maria Pedraza
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain; (M.P.); (V.M.M.)
| | - Victoria Moreno Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain; (M.P.); (V.M.M.)
| | - Laura Caramazza
- BioEMLab Group, DIET, Department of Information Engineering, Electronics and Telecommunications Sapienza, University of Rome, 00184 Rome, Italy; (L.C.); (M.C.); (F.A.); (M.L.)
| | - Micol Colella
- BioEMLab Group, DIET, Department of Information Engineering, Electronics and Telecommunications Sapienza, University of Rome, 00184 Rome, Italy; (L.C.); (M.C.); (F.A.); (M.L.)
| | | | | | - Francesca Apollonio
- BioEMLab Group, DIET, Department of Information Engineering, Electronics and Telecommunications Sapienza, University of Rome, 00184 Rome, Italy; (L.C.); (M.C.); (F.A.); (M.L.)
| | - Micaela Liberti
- BioEMLab Group, DIET, Department of Information Engineering, Electronics and Telecommunications Sapienza, University of Rome, 00184 Rome, Italy; (L.C.); (M.C.); (F.A.); (M.L.)
| | - Claudia Consales
- Division of Biotechnologies, Italian National Agency for Energy, New Technologies and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (G.B.); (C.C.); (V.P.); (F.C.); (C.M.)
| |
Collapse
|
3
|
Takaoka S, Uchida F, Ishikawa H, Toyomura J, Ohyama A, Matsumura H, Hirata K, Fukuzawa S, Kanno NI, Marushima A, Yamagata K, Yanagawa T, Matsumaru Y, Ishikawa E, Bukawa H. Sequencing-based study of neural induction of human dental pulp stem cells. Hum Cell 2024; 37:1638-1648. [PMID: 39210197 DOI: 10.1007/s13577-024-01121-7] [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: 06/13/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024]
Abstract
Techniques for triggering neural differentiation of embryonic and induced pluripotent stem cells into neural stem cells and neurons have been established. However, neural induction of mesenchymal stem cells, including dental pulp stem cells (DPSCs), has been assessed primarily based on neural-related gene regulation, and detailed studies into the characteristics and differentiation status of cells are lacking. Therefore, this study was aimed at evaluating the cellular components and differentiation pathways of neural lineage cells obtained via neural induction of human DPSCs. Human DPSCs were induced to neural cells in monolayer culture and examined for gene expression and mechanisms underlying differentiation using microarray-based ingenuity pathway analysis. In addition, the neural lineage cells were subjected to single-cell RNA sequencing (scRNA-seq) to classify cell populations based on gene expression profiles and to elucidate their differentiation pathways. Ingenuity pathway analysis revealed that genes exhibiting marked overexpression, post-neuronal induction, such as FABP7 and ZIC1, were associated with neurogenesis. Furthermore, in canonical pathway analysis, axon guidance signals demonstrated maximum activation. The scRNA-seq and cell type annotations revealed the presence of neural progenitor cells, astrocytes, neurons, and a small number of non-neural lineage cells. Moreover, trajectory and pseudotime analyses demonstrated that the neural progenitor cells initially engendered neurons, which subsequently differentiated into astrocytes. This result indicates that the aforementioned neural induction strategy generated neural stem/progenitor cells from DPSCs, which might differentiate and proliferate to constitute neural lineage cells. Therefore, neural induction of DPSCs may present an alternative approach to pluripotent stem cell-based therapeutic interventions for nervous system disorders.
Collapse
Affiliation(s)
- Shohei Takaoka
- Department of Oral and Maxillofacial Surgery, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Fumihiko Uchida
- Department of Oral and Maxillofacial Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junko Toyomura
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiro Ohyama
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hideaki Matsumura
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Koji Hirata
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoshi Fukuzawa
- Department of Oral and Maxillofacial Surgery, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Naomi Ishibashi Kanno
- Department of Oral and Maxillofacial Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aiki Marushima
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kenji Yamagata
- Department of Oral and Maxillofacial Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuji Matsumaru
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Eiichi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroki Bukawa
- Department of Oral and Maxillofacial Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
4
|
Hughes MP, Clarke KSP, Hoque R, Griffiths OV, Kruchek EJ, Johnson MP, Tariq MH, Kohli N, Lewis R, Labeed FH. Label-free, non-contact determination of resting membrane potential using dielectrophoresis. Sci Rep 2024; 14:18477. [PMID: 39122771 PMCID: PMC11316104 DOI: 10.1038/s41598-024-69000-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Measurement of cellular resting membrane potential (RMP) is important in understanding ion channels and their role in regulation of cell function across a wide range of cell types. However, methods available for the measurement of RMP (including patch clamp, microelectrodes, and potential-sensitive fluorophores) are expensive, slow, open to operator bias, and often result in cell destruction. We present non-contact, label-free membrane potential estimation which uses dielectrophoresis to determine the cytoplasm conductivity slope as a function of medium conductivity. By comparing this to patch clamp data available in the literature, we have demonstratet the accuracy of this approach using seven different cell types, including primary suspension cells (red blood cells, platelets), cultured suspension cells (THP-1), primary adherent cells (chondrocytes, human umbilical mesenchymal stem cells), and adherent (HeLa) and suspension (Jurkat) cancer cell lines. Analysis of the effect of ion channel inhibitors suggests the effects of pharmaceutical agents (TEA on HeLa; DMSO and neuraminidase on red blood cells) can also be measured. Comparison with published values of membrane potential suggest that the differences between our estimates and values recorded by patch clamp are accurate to within published margins of error. The method is low-cost, non-destructive, operator-independent and label-free, and has previously been shown to allow cells to be recovered after measurement.
Collapse
Affiliation(s)
- Michael Pycraft Hughes
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE.
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, UAE.
| | - Krista S P Clarke
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Rashedul Hoque
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Oreoluwa V Griffiths
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Emily J Kruchek
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Matthew P Johnson
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Muhammad Hamza Tariq
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Nupur Kohli
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Rebecca Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Fatima H Labeed
- Department of Biology, United Arab Emirates University, Al Ain, UAE.
| |
Collapse
|
5
|
Garrudo FFF, Linhardt RJ, Ferreira FC, Morgado J. Designing Electrical Stimulation Platforms for Neural Cell Cultivation Using Poly(aniline): Camphorsulfonic Acid. Polymers (Basel) 2023; 15:2674. [PMID: 37376320 DOI: 10.3390/polym15122674] [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: 04/19/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Electrical stimulation is a powerful strategy to improve the differentiation of neural stem cells into neurons. Such an approach can be implemented, in association with biomaterials and nanotechnology, for the development of new therapies for neurological diseases, including direct cell transplantation and the development of platforms for drug screening and disease progression evaluation. Poly(aniline):camphorsulfonic acid (PANI:CSA) is one of the most well-studied electroconductive polymers, capable of directing an externally applied electrical field to neural cells in culture. There are several examples in the literature on the development of PANI:CSA-based scaffolds and platforms for electrical stimulation, but no review has examined the fundamentals and physico-chemical determinants of PANI:CSA for the design of platforms for electrical stimulation. This review evaluates the current literature regarding the application of electrical stimulation to neural cells, specifically reviewing: (1) the fundamentals of bioelectricity and electrical stimulation; (2) the use of PANI:CSA-based systems for electrical stimulation of cell cultures; and (3) the development of scaffolds and setups to support the electrical stimulation of cells. Throughout this work, we critically evaluate the revised literature and provide a steppingstone for the clinical application of the electrical stimulation of cells using electroconductive PANI:CSA platforms/scaffolds.
Collapse
Affiliation(s)
- Fábio F F Garrudo
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Biology and Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Frederico Castelo Ferreira
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Jorge Morgado
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
6
|
Alqahtani S, Butcher MC, Ramage G, Dalby MJ, McLean W, Nile CJ. Acetylcholine Receptors in Mesenchymal Stem Cells. Stem Cells Dev 2023; 32:47-59. [PMID: 36355611 DOI: 10.1089/scd.2022.0201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are well known for their regenerative potential. Even though the ability of MSCs to proliferate and differentiate has been studied extensively, there remains much to learn about the signaling mechanisms and pathways that control proliferation and influence the differentiation phenotype. In recent years, there has been growing evidence for the utility of non-neuronal cholinergic signaling systems and that acetylcholine (ACh) plays an important ubiquitous role in cell-to-cell communication. Indeed, cholinergic signaling is hypothesized to occur in stem cells and ACh synthesis, as well as in ACh receptor (AChR) expression, has been identified in several stem cell populations, including MSCs. Furthermore, AChRs have been found to influence MSC regenerative potential. In humans, there are two major classes of AChRs, muscarinic AChRs and nicotinic AChRs, with each class possessing several subtypes or subunits. In this review, the expression and function of AChRs in different types of MSC are summarized with the aim of highlighting how AChRs play a pivotal role in regulating MSC regenerative function.
Collapse
Affiliation(s)
- Saeed Alqahtani
- School of Medicine Dentistry and Nursing and University of Glasgow, Glasgow, United Kingdom
| | - Mark C Butcher
- School of Medicine Dentistry and Nursing and University of Glasgow, Glasgow, United Kingdom
| | - Gordon Ramage
- School of Medicine Dentistry and Nursing and University of Glasgow, Glasgow, United Kingdom
| | - Matthew J Dalby
- School of Molecular Biosciences, University of Glasgow, Glasgow, United Kingdom
| | - William McLean
- School of Medicine Dentistry and Nursing and University of Glasgow, Glasgow, United Kingdom
| | - Christopher J Nile
- Faculty of Medical Sciences, School of Dental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
7
|
Fu Y, Ju Y, Zhao S. Ca v1.2 regulated odontogenic differentiation of NG2 + pericytes during pulp injury. Odontology 2023; 111:57-67. [PMID: 35739380 DOI: 10.1007/s10266-022-00720-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/30/2022] [Indexed: 01/06/2023]
Abstract
NG2+ pericytes, as the possible precursor cells of mesenchymal stem cells (MSCs), have drawn attention due to their ability to differentiate into odontoblasts. Cav1.2 is involved in the differentiation process of stem cells, but its role in the differentiation of NG2+ pericytes is not clear. The aim of the present study was to examine the role of Cav1.2 in the differentiation of NG2+ pericytes into odontoblasts. NG2+ pericytes were obtained from human dental pulp cells by magnetic-activated cell sorting. During the odontogenic differentiation of NG2+ pericytes, the effects of the Cav1.2 inhibitors, nimodipine and Cav1.2 knockdown shRNA, were analyzed by real-time polymerase chain reaction and alizarin red staining. NG2CreERT2/Rosa26-GFP lineage-tracing mice were established to further investigate the roles of NG2+ pericytes and Cav1.2 in incisor self-repair after injury in vivo. At 10 min, 1 day, and 3 days after pulp injuries in transgenic mice, NG2-GFP+ and Cav1.2 immunofluorescence co-staining was performed on the incisors. Nimodipine treatment and Cav1.2 knockdown showed similar inhibition of calcium nodule formation and mRNA levels of osteogenic markers (DSPP, DMP1, and Runx2, p < 0.05). NG2+ pericytes migrated from their inherent perivascular location to the odontoblast layers after pulp injury. Cav1.2 showed a similar response pattern as NG2+ pericytes and gradually returned to normal levels. In addition, many co-stained areas of Cav1.2 and NG2+ pericytes, both near the perivascular and odontoblast layers, were observed. These results indicate that Cav1.2 played a vital role in the odontogenic differentiation of NG2+ pericytes, and that it might be closely linked to the NG2+ pericytes-mediated repair of dental pulp injury in vivo.
Collapse
Affiliation(s)
- Yunyu Fu
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Yanqin Ju
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Shouliang Zhao
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
8
|
Potier-Cartereau M, Gautier M, Ravalet N, Ducrocq E, Hamard S, LeGuennec JY, Vandier C, Herault O. The Sodium-Calcium Exchanger Controls the Membrane Potential of AFT024: A Mesenchymal Stem Cell Hematopoietic Niche Forming Line. Bioelectricity 2022; 4:103-107. [PMID: 39350778 PMCID: PMC11441364 DOI: 10.1089/bioe.2022.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to characterize the functional expression of sodium-calcium exchangers on AFT024 cell line, a murine model of mesenchymal stem/stromal cells (MSCs) supporting human primitive hematopoiesis. All current-clamp and voltage-clamp experiments were performed using the perforated patch whole-cell recording technique with amphotericin B. The membrane potential of -14 mV shifted to -35 mV when lowering the external sodium concentration to 0.33 mM and an increase of cytosolic calcium concentration was observed. KB-R7943, a selective blocker of cardiac sodium-calcium exchangers, also named NCX1, induced a hyperpolarization at physiological sodium concentration while it blocked the hyperpolarization observed at low sodium concentration. This demonstrates for the first time the presence of the sodium-calcium exchangers in AFT024 cells and provides initial evidence that the membrane potential of these stromal cells is maintained depolarized by this exchanger. Lowering external sodium concentration and KB-R7943 had no effect on the membrane potential of 2018 cells, a nonhematopoietic-supportive cell line. Since NCX1 is differentially expressed in AFT024 cells as compared with nonhematopoietic supportive cells with more restricted differentiation potential, this study suggests a potential role of this sodium-calcium exchanger, in the differentiation process or hematopoietic support of MSCs.
Collapse
Affiliation(s)
| | - Mathieu Gautier
- LPCM UR 4667, University of Picardie Jules Verne, Amiens, France
| | - Noemie Ravalet
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
| | - Elfi Ducrocq
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
| | - Sophie Hamard
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
| | - Jean-Yves LeGuennec
- UMR 01046–UMR 9214, University of Montpellier, INSERM, CNRS, Montpellier, France
| | | | - Olivier Herault
- LNOx EMR 7001/EA 7501, University of Tours, CNRS, Tours, France
- Department of Biological Hematology, Tours University Hospital, Tours, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
9
|
Talmon M, Massara E, Pruonto G, Quaregna M, Boccafoschi F, Riva B, Fresu LG. Characterization of a functional Ca2+ toolkit in urine-derived stem cells and derived skeletal muscle cells. Cell Calcium 2022; 103:102548. [DOI: 10.1016/j.ceca.2022.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/11/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022]
|
10
|
Shenfu Injection: A Famous Chinese Prescription That Promotes HCN4 Activity in Bone Marrow Mesenchymal Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9912844. [PMID: 34457032 PMCID: PMC8387162 DOI: 10.1155/2021/9912844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/27/2021] [Accepted: 08/08/2021] [Indexed: 11/30/2022]
Abstract
We investigated the effects of Shenfu Injection (SFI) on HCN4 activity in bone marrow mesenchymal stem cells (BMSCs). The sample of BMSCs was divided into six groups: a control group, a high-dose SFI group (0.25 ml/ml), a middle-dose SFI group (0.1 ml/ml), a low-dose SFI group (0.05 ml/ml), an adenovirus-encoded control vector group, and an adenovirus-encoded HCN4 group. Cell ultrastructure was observed using a transmission electron microscope. Quantitative reverse transcription PCR (RT-qPCR) was performed to detect HCN4 expression, and HCN4 activity was detected using the whole-cell patch clamp technique. An enzyme-linked immunosorbent assay was performed to detect cAMP content. Application of flow cytometry confirmed that the isolated cells showed BMSC-like phenotypes. Differentiation of BMSCs in both the SFI and the adenovirus-encoding HCN4 groups occurred according to the cellular ultrastructure. Application of the whole-cell patch clamp technique revealed that SFI could activate the inward pacing current of BMSCs in a concentration-dependent manner. The RT-qPCR results showed that HCN4 expression was significantly higher in the high-dose SFI group than in the medium- and low-dose groups, whereas the cAMP content in the overexpressed HCN4 group decreased significantly; this content in the high-dose SFI group increased significantly. In conclusion, SFI promotes HCN4 activity in BMSCs, which could explain its treatment effect when administered to patients with cardiovascular diseases.
Collapse
|
11
|
Janus 3D printed dynamic scaffolds for nanovibration-driven bone regeneration. Nat Commun 2021; 12:1031. [PMID: 33589620 PMCID: PMC7884435 DOI: 10.1038/s41467-021-21325-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
The application of physical stimuli to cell cultures has shown potential to modulate multiple cellular functions including migration, differentiation and survival. However, the relevance of these in vitro models to future potential extrapolation in vivo depends on whether stimuli can be applied “externally”, without invasive procedures. Here, we report on the fabrication and exploitation of dynamic additive-manufactured Janus scaffolds that are activated on-command via external application of ultrasounds, resulting in a mechanical nanovibration that is transmitted to the surrounding cells. Janus scaffolds were spontaneously formed via phase-segregation of biodegradable polycaprolactone (PCL) and polylactide (PLA) blends during the manufacturing process and behave as ultrasound transducers (acoustic to mechanical) where the PLA and PCL phases represent the active and backing materials, respectively. Remote stimulation of Janus scaffolds led to enhanced cell proliferation, matrix deposition and osteogenic differentiation of seeded human bone marrow derived stromal cells (hBMSCs) via formation and activation of voltage-gated calcium ion channels. Fabrication of dynamic, reversible and biocompatible scaffolds with non-invasive external triggers has so far been limited. Here, the authors report on the creation of 3D printed scaffolds with Janus structure that produce nanovibrations when exposed to ultrasound, promoting bone regeneration.
Collapse
|
12
|
Henao JC, Grismaldo A, Barreto A, Rodríguez-Pardo VM, Mejía-Cruz CC, Leal-Garcia E, Pérez-Núñez R, Rojas P, Latorre R, Carvacho I, Torres YP. TRPM8 Channel Promotes the Osteogenic Differentiation in Human Bone Marrow Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:592946. [PMID: 33614639 PMCID: PMC7890257 DOI: 10.3389/fcell.2021.592946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/05/2021] [Indexed: 11/29/2022] Open
Abstract
Various families of ion channels have been characterized in mesenchymal stem cells (MSCs), including some members of transient receptor potential (TRP) channels family. TRP channels are involved in critical cellular processes as differentiation and cell proliferation. Here, we analyzed the expression of TRPM8 channel in human bone marrow MSCs (hBM-MSCs), and its relation with osteogenic differentiation. Patch-clamp recordings showed that hBM-MSCs expressed outwardly rectifying currents which were increased by exposure to 500 μM menthol and were partially inhibited by 10 μM of BCTC, a TRPM8 channels antagonist. Additionally, we have found the expression of TRPM8 by RT-PCR and western blot. We also explored the TRPM8 localization in hBM-MSCs by immunofluorescence using confocal microscopy. Remarkably, hBM-MSCs treatment with 100 μM of menthol or 10 μM of icilin, TRPM8 agonists, increases osteogenic differentiation. Conversely, 20 μM of BCTC, induced a decrease of osteogenic differentiation. These results suggest that TRPM8 channels are functionally active in hBM-MSCs and have a role in cell differentiation.
Collapse
Affiliation(s)
- Juan C Henao
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Grismaldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Viviana M Rodríguez-Pardo
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Claudia Camila Mejía-Cruz
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Efrain Leal-Garcia
- Departamento de Ortopedia y Traumatología, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Patricio Rojas
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
13
|
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
|
14
|
Kachroo U, Livingston A, Vinod E, Sathishkumar S, Boopalan PRJVC. Comparison of Electrophysiological Properties and Gene Expression between Human Chondrocytes and Chondroprogenitors Derived from Normal and Osteoarthritic Cartilage. Cartilage 2020; 11:374-384. [PMID: 30139266 PMCID: PMC7298598 DOI: 10.1177/1947603518796140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES Bone-marrow mesenchymal stem cells (MSCs) and chondrocytes are currently used for cell-based therapy in cartilage repair. Chondroprogenitors (CPs), resident cells of articular cartilage, demonstrate likeness to stem cells. Reports suggest that chondrocytes phenotype is altered in culture, thus making differentiation between the two cell populations difficult. Our objectives were to electrophysiologically assess chondrocytes and CPs, compare their mRNA expression with that of ionic channels already reported in MSCs, and to observe the effect of time in culture and osteoarthritic damage on cells. DESIGN AND RESULTS Chondrocytes and CPs at passages 0 (p0) and 5 (p5) derived from normal and osteoarthritic (OA) knee joints were used. Ionic currents were recorded by subjecting cells to depolarizing voltage pulses, and reverse transcriptase-polymerase chain reaction (RT-PCR) was used for studying ion channel expression. Our results demonstrated that both chondrocytes and CPs showed the presence of similar currents belonging to voltage-gated potassium channel subfamily, with RT-PCR confirming high mRNA expression of Maxi K, HKv1.1, HKv1.4, HKv4.2, and hEAG1 channels. Our finding also suggested that CPs were comparatively more sensitive to increased time in culture and inflammatory processes as observed in OA, as was evidenced by the significant decrease in mean current density (p0 normal CP: 183.171 ± 50.80 pA/pF; p5 normal CP: 50.225 ± 17.63 pA/pF; P = 0.0280) and significant increase in cellular size (p0 normal CP: 21.564 ± 2.98 pF; p0 OA CP: 37.939 ± 3.55 pF; P = 0.0057). CONCLUSION Both cell types appear to be optimal candidates for cell-based therapy although initial seeding density, cell source (normal vs. OA), and time in culture are matters of concern, prior to cell-type selection.
Collapse
Affiliation(s)
- Upasana Kachroo
- Department of Physiology, Christian
Medical College, Vellore, India
| | - Abel Livingston
- Department of Orthopaedics, Christian
Medical College, Vellore, India
| | - Elizabeth Vinod
- Department of Physiology, Christian
Medical College, Vellore, India,Centre for Stem Cell Research, Christian
Medical College, Vellore, India,Elizabeth Vinod, Department of Physiology,
Christian Medical College, Bagayam, Vellore 632002, Tamil Nadu, India.
| | | | - P. R. J. V. C. Boopalan
- Department of Orthopaedics, Christian
Medical College, Vellore, India,Centre for Stem Cell Research, Christian
Medical College, Vellore, India
| |
Collapse
|
15
|
The voltage-gated proton channel hHv1 is functionally expressed in human chorion-derived mesenchymal stem cells. Sci Rep 2020; 10:7100. [PMID: 32346069 PMCID: PMC7188850 DOI: 10.1038/s41598-020-63517-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/01/2020] [Indexed: 01/08/2023] Open
Abstract
The voltage-gated proton channel Hv1 is widely expressed, among others, in immune and cancer cells, it provides an efficient cytosolic H+extrusion mechanism and regulates vital functions such as oxidative burst, migration and proliferation. Here we demonstrate the presence of human Hv1 (hHv1) in the placenta/chorion-derived mesenchymal stem cells (cMSCs) using RT-PCR. The voltage- and pH-dependent gating of the current is similar to that of hHv1 expressed in cell lines and that the current is blocked by 5-chloro-2-guanidinobenzimidazole (ClGBI) and activated by arachidonic acid (AA). Inhibition of hHv1 by ClGBI significantly decreases mineral matrix production of cMSCs induced by conditions mimicking physiological or pathological (inorganic phosphate, Pi) induction of osteogenesis. Wound healing assay and single cell motility analysis show that ClGBI significantly inhibits the migration of cMSCs. Thus, seminal functions of cMSCs are modulated by hHv1 which makes this channel as an attractive target for controlling advantages/disadvantages of MSCs therapy.
Collapse
|
16
|
Echeverry S, Grismaldo A, Sánchez C, Sierra C, Henao JC, Granados ST, Sutachán JJ, Torres YP. Activation of BK Channel Contributes to PL-Induced Mesenchymal Stem Cell Migration. Front Physiol 2020; 11:210. [PMID: 32265729 PMCID: PMC7105713 DOI: 10.3389/fphys.2020.00210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/24/2020] [Indexed: 01/16/2023] Open
Abstract
Due to their capacity to proliferate, migrate, and differentiate, mesenchymal stem cells (MSCs) are considered to be good candidates for regenerative medicine applications. The mechanisms underlying proliferation and differentiation of MSCs have been studied. However, much less is known about the mechanisms regulating the migration of MSCs. Platelet lysate (PL), a supplement used to promote cell expansion, has been shown to promote MSCs migration; however, the underlying mechanism are unknown. Here, by using adipose-derived rat MSCs (rMSCs) and the scratch assay in the absence and presence of various BK channels modulators, we evaluated the role of BK channels in mediating the PL-stimulated migration of rMSCs. We found that 5% PL increased rMSCs migration, and this effect was blocked by the addition of the BK channel selective antagonist Iberiotoxin (IBTX). In the absence of PL, the BK channel agonist NS1619, stimulated rMSCs migration to similar level as 5% PL. Addition of both NS1619 and 5% PL resulted in an increase in rMSCs migration, that was higher than when either one was added individually. From whole-cell recordings, it was found that the addition of 5% PL increased the magnitude of BK current density. By using Western blot and flow cytometry, it was found that PL did not affect the expression of BK channels. Together, our results indicate that as shown in other cell types, activation of BK channels by themselves also promote rMSC migration, and show that activation of BK channels contribute to the observed PL-induced increase in migration of rMSC.
Collapse
|
17
|
Primary Cilia Mediate Wnt5a/β-catenin Signaling to Regulate Adipogenic Differentiation of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Following Calcium Induction. Tissue Eng Regen Med 2020; 17:193-202. [PMID: 32008170 DOI: 10.1007/s13770-019-00237-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/17/2019] [Accepted: 12/22/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Regeneration of soft tissue defects is essential for adipose tissue pathologies and disease, trauma, or injury-induced damage. Here, we show that umbilical cord blood-derived mesenchymal stem cells could potentially be tailored and used for the reconstruction of specific damaged sites. Adipogenesis can be exploited in soft tissue reconstruction. Also, primary cilia play a role in the control of adipogenesis. METHODS The adipogenic differentiation capacity of mesenchymal stem cells (MSCs) was shown to influence ciliogenesis. MSCs transfected with intraflagellar transport 88 (IFT88) small interfering RNA (siRNA), which blocks the assembly and maintenance of cilia, were examined to confirm the relationship between adipogenesis and ciliogenesis. Also, 1,2-Bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA-AM), calcium chelator, inhibited the ciliogenesis of MSCs in adipogenic differentiation. RESULTS IFT88-knockdown led to decreased cilia formation and limitation of cilia elongation in adipogenesis. Additionally, intracellular calcium triggered cilia formation in MSCs adipogenesis. Interestingly, intracellular calcium cannot overcome the inhibition of adipogenesis caused by low numbers of cilia in MSCs. CONCLUSION Our data suggested that ciliogenesis was negatively regulated by Wnt5a/β-catenin signaling during adipogenesis. Thus, we suggest that calcium induction triggers adipogenesis and ciliogenesis.
Collapse
|
18
|
Chen K, Huang Y, Singh R, Wang ZZ. Arrhythmogenic risks of stem cell replacement therapy for cardiovascular diseases. J Cell Physiol 2020; 235:6257-6267. [PMID: 31994198 DOI: 10.1002/jcp.29554] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Ischemic heart disease and congestive heart failure are major contributors to high morbidity and mortality. Approximately 1.5 million cases of myocardial infarction occur annually in the United States; the yearly incidence rate is approximately 600 cases per 100,000 people. Although significant progress to improve the survival rate has been made by medications and implantable medical devices, damaged cardiomyocytes are unable to be recovered by current treatment strategies. After almost two decades of research, stem cell therapy has become a very promising approach to generate new cardiomyocytes and enhance the function of the heart. Along with clinical trials with stem cells conducted in cardiac regeneration, concerns regarding safety and potential risks have emerged. One of the contentious issues is the electrical dysfunctions of cardiomyocytes and cardiac arrhythmia after stem cell therapy. In this review, we focus on the cell sources currently used for stem cell therapy and discuss related arrhythmogenic risk.
Collapse
Affiliation(s)
- Kang Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Huang
- Department of Medicine, University of Maryland Medical Center Midtown Campus, Baltimore, Maryland
| | - Radhika Singh
- Center for Biotechnology Education, Johns Hopkins University, Baltimore, Maryland
| | - Zack Z Wang
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Case Report on: Very Early Afterdepolarizations in HiPSC-Cardiomyocytes-An Artifact by Big Conductance Calcium Activated Potassium Current (I bk,Ca). Cells 2020; 9:cells9010253. [PMID: 31968557 PMCID: PMC7017352 DOI: 10.3390/cells9010253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/16/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) represent an unlimited source of human CMs that could be a standard tool in drug research. However, there is concern whether hiPSC-CMs express all cardiac ion channels at physiological level and whether they might express non-cardiac ion channels. In a control hiPSC line, we found large, “noisy” outward K+ currents, when we measured outward potassium currents in isolated hiPSC-CMs. Currents were sensitive to iberiotoxin, the selective blocker of big conductance Ca2+-activated K+ current (IBK,Ca). Seven of 16 individual differentiation batches showed a strong initial repolarization in the action potentials (AP) recorded from engineered heart tissue (EHT) followed by very early afterdepolarizations, sometimes even with consecutive oscillations. Iberiotoxin stopped oscillations and normalized AP shape, but had no effect in other EHTs without oscillations or in human left ventricular tissue (LV). Expression levels of the alpha-subunit (KCa1.1) of the BKCa correlated with the presence of oscillations in hiPSC-CMs and was not detectable in LV. Taken together, individual batches of hiPSC-CMs can express sarcolemmal ion channels that are otherwise not found in the human heart, resulting in oscillating afterdepolarizations in the AP. HiPSC-CMs should be screened for expression of non-cardiac ion channels before being applied to drug research.
Collapse
|
20
|
BK ablation attenuates osteoblast bone formation via integrin pathway. Cell Death Dis 2019; 10:738. [PMID: 31570694 PMCID: PMC6769012 DOI: 10.1038/s41419-019-1972-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 11/09/2022]
Abstract
Impaired bone formation is one of the major causes of low bone mass and skeletal fragility that occurs in osteoporosis. However, the mechanisms underlying the defects in bone formation are not well understood. Here, we report that big conductance calcium-activated potassium channels (BKs) are required for bone formation and osteoblast function both in vivo and in vitro. By 15 weeks of age, BK knockout (BKO) mice exhibited a decline in bone mineral density and trabecular bone volume of the tibiae and lumbar vertebrae, which were associated with impaired bone formation and osteoblast activity. Mechanistically, BK ablation in bone and bone marrow mesenchymal stem cells (BMSCs) of BKO mice inhibited integrin signaling. Furthermore, the binding of α subunit of BK with integrin β1 protein in osteoblasts was confirmed, and FAK-ERK1/2 signaling was proved to be involved by genetic modification of KCNMA1 (which encodes the α subunit of BK) in ROS17/2.8 osteoblast cells. These findings indicated that BK regulates bone formation by promoting osteoblast differentiation via integrin pathway, which provided novel insight into ion transporter crosstalk with the extracellular matrix in osteoblast regulation and revealed a new potential strategy for intervention in correcting bone formation defects.
Collapse
|
21
|
Alfieri R, Vassalli M, Viti F. Flow-induced mechanotransduction in skeletal cells. Biophys Rev 2019; 11:729-743. [PMID: 31529361 DOI: 10.1007/s12551-019-00596-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Human body is subject to many and variegated mechanical stimuli, actuated in different ranges of force, frequency, and duration. The process through which cells "feel" forces and convert them into biochemical cascades is called mechanotransduction. In this review, the effects of fluid shear stress on bone cells will be presented. After an introduction to present the major players in bone system, we describe the mechanoreceptors in bone tissue that can feel and process fluid flow. In the second part of the review, we present an overview of the biological processes and biochemical cascades initiated by fluid shear stress in bone cells.
Collapse
Affiliation(s)
- Roberta Alfieri
- Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" - National Research Council (IGM-CNR), Via Abbiategrasso, 207, 27100, Pavia, Italy
| | - Massimo Vassalli
- Institute of Biophysics - National Research Council (IBF-CNR), Via De Marini, 6, 16149, Genoa, Italy
| | - Federica Viti
- Institute of Biophysics - National Research Council (IBF-CNR), Via De Marini, 6, 16149, Genoa, Italy.
| |
Collapse
|
22
|
Silva HB, Rodrigues DC, Andrade R, Teixeira GHGSF, Stelling MP, Ponte CG, Nascimento JHM, Campos de Carvalho AC, Medei E. Expression of potassium channels is relevant for cell survival and migration in a murine bone marrow stromal cell line. J Cell Physiol 2019; 234:18086-18097. [PMID: 30887515 DOI: 10.1002/jcp.28441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 02/05/2023]
Abstract
S17 is a clonogenic bone marrow stromal (BMS) cell line derived from mouse that has been extensively used to assess both human and murine hematopoiesis support capacity. However, very little is known about the expression of potassium ion channels and their function in cell survival and migration in these cells. Thus, the present study was designed to characterize potassium ion channels using electrophysiological and molecular biological approaches in S17 BMS cells. The whole-cell configuration of the patch clamp technique has been applied to identify potassium ion currents and reverse transcription polymerase chain reaction (RT-PCR) used to determine their molecular identities. Based on gating kinetics and pharmacological modulation of the macroscopic currents we found the presence of four functional potassium ion channels in S17 BMS cells. These include a current rapidly activated and inactivated, tetraethylammonium-sensitive, (IKV ) in most (50%) cells; a fast activated and rapidly inactivating A-type K + current (IK A -like); a delayed rectifier K + current (IK DR ) and an inward rectifier potassium current (IK IR ), found in, respectively 4.5%, 26% and 24% of these cells. RT-PCR confirmed the presence of mRNA transcripts for the alpha subunit of the corresponding functional ion channels. Additionally, functional assays were performed to investigate the importance of potassium currents in cell survival and migration. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analyses revealed a reduction in cell viability, while wound healing assays revealed reduced migration potential in cells incubated with different potassium channel blockers. In conclusion, our data suggested that potassium currents might play a role in the maintenance of overall S17 cell ionic homeostasis directly affecting cell survival and migration.
Collapse
Affiliation(s)
- Henrique B Silva
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Center for Neuroscience and Cell Biology, University of Coimbra Rua Larga, Faculty of Medicine, Coimbra, Portugal
| | - Deivid C Rodrigues
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Raiana Andrade
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriel H G S F Teixeira
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana P Stelling
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Cristiano G Ponte
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José H M Nascimento
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Emiliano Medei
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
23
|
Darche FF, Rivinius R, Köllensperger E, Leimer U, Germann G, Seckinger A, Hose D, Schröter J, Bruehl C, Draguhn A, Gabriel R, Schmidt M, Koenen M, Thomas D, Katus HA, Schweizer PA. Pacemaker cell characteristics of differentiated and HCN4-transduced human mesenchymal stem cells. Life Sci 2019; 232:116620. [PMID: 31291594 DOI: 10.1016/j.lfs.2019.116620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/21/2019] [Accepted: 06/29/2019] [Indexed: 12/13/2022]
Abstract
AIMS Cell-based biological pacemakers aim to overcome limitations and side effects of electronic pacemaker devices. We here developed and tested different approaches to achieve nodal-type differentiation using human adipose- and bone marrow-derived mesenchymal stem cells (haMSC, hbMSC). MAIN METHODS haMSC and hbMSC were differentiated using customized protocols. Quantitative RT-PCR was applied for transcriptional pacemaker-gene profiling. Protein membrane expression was analyzed by immunocytochemistry. Pacemaker current (If) was studied in haMSC with and without lentiviral HCN4-transduction using patch clamp recordings. Functional characteristics were evaluated by co-culturing with neonatal rat ventricular myocytes (NRVM). KEY FINDINGS Culture media-based differentiation for two weeks generated cells with abundant transcription of ion channel genes (Cav1.2, NCX1), transcription factors (TBX3, TBX18, SHOX2) and connexins (Cx31.9 and Cx45) characteristic for cardiac pacemaker tissue, but lack adequate HCN transcription. haMSC-derived cells revealed transcript levels, which were closer related to sinoatrial nodal cells than hbMSC-derived cells. To substitute for the lack of If, we performed lentiviral HCN4-transduction of haMSC resulting in stable If. Co-culturing with NRVM demonstrated that differentiated haMSC expressing HCN4 showed earlier onset of spontaneous contractions and higher beating regularity, synchrony and rate compared to co-cultures with non-HCN4-transduced haMSC or HCN4-transduced, non-differentiated haMSC. Confocal imaging indicated increased membrane expression of cardiac gap junctional proteins in differentiated haMSC. SIGNIFICANCE By differentiation haMSC, rather than hbMSC attain properties favorable for cardiac pacemaking. In combination with lentiviral HCN4-transduction, a cellular phenotype was generated that sustainably controls and stabilizes rate in co-culture with NRVM.
Collapse
Affiliation(s)
- Fabrice F Darche
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Rasmus Rivinius
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Eva Köllensperger
- ETHIANUM Klinik Heidelberg, Voßstraße 6, D-69115 Heidelberg, Germany
| | - Uwe Leimer
- ETHIANUM Klinik Heidelberg, Voßstraße 6, D-69115 Heidelberg, Germany
| | - Günter Germann
- ETHIANUM Klinik Heidelberg, Voßstraße 6, D-69115 Heidelberg, Germany
| | - Anja Seckinger
- Department of Hematology, Oncology and Rheumatology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Dirk Hose
- Department of Hematology, Oncology and Rheumatology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Julian Schröter
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Claus Bruehl
- Institute for Physiology and Pathophysiology, University of Heidelberg, INF 326, D-69120 Heidelberg, Germany
| | - Andreas Draguhn
- Institute for Physiology and Pathophysiology, University of Heidelberg, INF 326, D-69120 Heidelberg, Germany
| | - Richard Gabriel
- Molecular and Gene Therapy, National Center for Tumor Diseases (NCT) Heidelberg, INF 460, D-69120 Heidelberg, Germany
| | - Manfred Schmidt
- Molecular and Gene Therapy, National Center for Tumor Diseases (NCT) Heidelberg, INF 460, D-69120 Heidelberg, Germany
| | - Michael Koenen
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany; Department of Molecular Neurobiology, Max-Planck-Institute for Medical Research, Jahnstrasse 29, D-69120 Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120 Heidelberg, Germany
| | - Patrick A Schweizer
- Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120 Heidelberg, Germany.
| |
Collapse
|
24
|
Tan Y, Fei D, He X, Dai J, Xu R, Xu X, Wu J, Li B. L-type voltage-gated calcium channels in stem cells and tissue engineering. Cell Prolif 2019; 52:e12623. [PMID: 31115100 PMCID: PMC6669004 DOI: 10.1111/cpr.12623] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 12/22/2022] Open
Abstract
L-type voltage-gated calcium ion channels (L-VGCCs) have been demonstrated to be the mediator of several significant intracellular activities in excitable cells, such as neurons, chromaffin cells and myocytes. Recently, an increasing number of studies have investigated the function of L-VGCCs in non-excitable cells, particularly stem cells. However, there appear to be no systematic reviews of the relationship between L-VGCCs and stem cells, and filling this gap is prescient considering the contribution of L-VGCCs to the proliferation and differentiation of several types of stem cells. This review will discuss the possible involvement of L-VGCCs in stem cells, mainly focusing on osteogenesis mediated by mesenchymal stem cells (MSCs) from different tissues and neurogenesis mediated by neural stem/progenitor cells (NSCs). Additionally, advanced applications that use these channels as the target for tissue engineering, which may offer the hope of tissue regeneration in the future, will also be explored.
Collapse
Affiliation(s)
- Yi‐zhou Tan
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Dong‐dong Fei
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| | - Xiao‐ning He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| | - Ji‐min Dai
- Doctoral students of eight-year programThe Fourth Military Medical UniversityXi’anChina
| | - Rong‐chen Xu
- Doctoral students of eight-year programThe Fourth Military Medical UniversityXi’anChina
| | - Xin‐yue Xu
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Jun‐jie Wu
- Department of Orthodontics, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Bei Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| |
Collapse
|
25
|
Molecular phenotyping of the surfaceome of migratory chondroprogenitors and mesenchymal stem cells using biotinylation, glycocapture and quantitative LC-MS/MS proteomic analysis. Sci Rep 2019; 9:9018. [PMID: 31227739 PMCID: PMC6588563 DOI: 10.1038/s41598-019-44957-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
The complement of cell surface proteins, collectively referred to as the surfaceome, is a useful indicator of normal differentiation processes, and the development of pathologies such as osteoarthritis (OA). We employed biochemical and proteomic tools to explore the surfaceome and to define biomarkers in chondrogenic progenitor cells (CPC) derived from human OA knee articular cartilage. These cells have great therapeutic potential, but their unexplored biology limits their clinical application. We performed biotinylation combined with glycocapture and high throughput shotgun proteomics to define the surface proteome of human bone marrow mesenchymal stem cells (MSCs) and human CPCs. We prepared cell surface protein-enriched fractions from MSCs and CPCs, and then a proteomic approach was used to compare and evaluate protein changes between undifferentiated MSCs and CPCs. 1256 proteins were identified in the study, of which 791 (63%) were plasma membrane, cell surface or extracellular matrix proteins. Proteins constituting the surfaceome were annotated and categorized. Our results provide, for the first time, a repository of quantitative proteomic data on the surfaceome of two closely related cell types relevant to cartilage biology and OA. These results may provide novel insights into the transformation of the surfaceome during chondrogenic differentiation and phenotypic changes during OA development.
Collapse
|
26
|
Abstract
Modern stem cell research has mainly focused on protein expression and transcriptional networks. However, transmembrane voltage gradients generated by ion channels and transporters have demonstrated to be powerful regulators of cellular processes. These physiological cues exert influence on cell behaviors ranging from differentiation and proliferation to migration and polarity. Bioelectric signaling is a fundamental element of living systems and an untapped reservoir for new discoveries. Dissecting these mechanisms will allow for novel methods of controlling cell fate and open up new opportunities in biomedicine. This review focuses on the role of ion channels and the resting membrane potential in the proliferation and differentiation of skeletal muscle progenitor cells. In addition, findings relevant to this topic are presented and potential implications for tissue engineering and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Colin Fennelly
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Inc., Cambridge, Massachusetts
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
27
|
Cheng Q, Chen A, Du Q, Liao Q, Shuai Z, Chen C, Yang X, Hu Y, Zhao J, Liu S, Wen GR, An J, Jing H, Tuo B, Xie R, Xu J. Novel insights into ion channels in cancer stem cells (Review). Int J Oncol 2018; 53:1435-1441. [PMID: 30066845 DOI: 10.3892/ijo.2018.4500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/28/2018] [Indexed: 11/06/2022] Open
Abstract
Cancer stem cells (CSCs) are immortal cells in tumor tissues that have been proposed as the driving force of tumorigenesis and tumor invasion. Previously, ion channels were revealed to contribute to cancer cell proliferation, migration and apoptosis. Recent studies have demonstrated that ion channels are present in various CSCs; however, the functions of ion channels and their mechanisms in CSCs remain unknown. The present review aimed to focus on the roles of ion channels in the regulation of CSC behavior and the CSC-like properties of cancer cells. Evaluation of the relationship between ion channels and CSCs is critically important for understanding malignancy.
Collapse
Affiliation(s)
- Qijiao Cheng
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Anhai Chen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Zhangli Shuai
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Changmei Chen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Xinrong Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Yaxia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Ju Zhao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Songpo Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Guo Rong Wen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Jiaxin An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Hai Jing
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
28
|
Intracellular Calcium Determines the Adipogenic Differentiation Potential of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells via the Wnt5a/ β-Catenin Signaling Pathway. Stem Cells Int 2018; 2018:6545071. [PMID: 30123291 PMCID: PMC6079381 DOI: 10.1155/2018/6545071] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/03/2018] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells- (MSCs-) based therapies show different degrees of efficacies for the treatment of various diseases, including lipogenesis. We evaluated the adipogenic differentiation ability of human umbilical cord blood-derived MSCs (hUCB-MSCs) from different donors and examined the contribution of the intracellular calcium (Ca2+) level to this diversity. hUCB-MSCs treated with Ca2+ or the Ca2+ chelator BAPTA-AM increased and decreased adipogenic differentiation, respectively. Canonical Wnt5a/β-catenin expression decreased during adipogenic differentiation of hUCB-MSCs. Treatment with Wnt5a blocked the adipogenic differentiation of hUCB-MSCs and activated the Wnt pathway, with a decrease in the adipogenesis markers PPARγ and leptin, and reduced lipid vacuole-associated Oil red O activity. In contrast, inhibition of the Wnt pathway with dickkopf-1 and β-catenin small interfering RNA transfection promoted the adipogenic potential of hUCB-MSCs. Interestingly, the Ca2+-based system exhibited a synergic effect on adipogenic potential through the Wnt5a/β-catenin pathway. Our data suggest that the variable adipogenic differentiation potential of hUCB-MSCs from different lots is due to variation in the intracellular Ca2+ level, which can be used as a marker to predict hUCB-MSCs selection for lipogenesis therapy. Overall, these results demonstrate that exogenous calcium treatment enhanced the adipogenic differentiation of hUCB-MSCs via negatively regulating the Wnt5a/β-catenin signaling pathway.
Collapse
|
29
|
Cortical and spinal conditioned media modify the inward ion currents and excitability and promote differentiation of human striatal primordium. J Chem Neuroanat 2018; 90:87-97. [DOI: 10.1016/j.jchemneu.2017.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 11/18/2022]
|
30
|
Khlusov IA, Dekhtyar Y, Sharkeev YP, Pichugin VF, Khlusova MY, Polyaka N, Tyulkin F, Vendinya V, Legostaeva EV, Litvinova LS, Shupletsova VV, Khaziakhmatova OG, Yurova KA, Prosolov KA. Nanoscale Electrical Potential and Roughness of a Calcium Phosphate Surface Promotes the Osteogenic Phenotype of Stromal Cells. MATERIALS 2018; 11:ma11060978. [PMID: 29890754 PMCID: PMC6024922 DOI: 10.3390/ma11060978] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells (MSCs) and osteoblasts respond to the surface electrical charge and topography of biomaterials. This work focuses on the connection between the roughness of calcium phosphate (CP) surfaces and their electrical potential (EP) at the micro- and nanoscales and the possible role of these parameters in jointly affecting human MSC osteogenic differentiation and maturation in vitro. A microarc CP coating was deposited on titanium substrates and characterized at the micro- and nanoscale. Human adult adipose-derived MSCs (hAMSCs) or prenatal stromal cells from the human lung (HLPSCs) were cultured on the CP surface to estimate MSC behavior. The roughness, nonuniform charge polarity, and EP of CP microarc coatings on a titanium substrate were shown to affect the osteogenic differentiation and maturation of hAMSCs and HLPSCs in vitro. The surface EP induced by the negative charge increased with increasing surface roughness at the microscale. The surface relief at the nanoscale had an impact on the sign of the EP. Negative electrical charges were mainly located within the micro- and nanosockets of the coating surface, whereas positive charges were detected predominantly at the nanorelief peaks. HLPSCs located in the sockets of the CP surface expressed the osteoblastic markers osteocalcin and alkaline phosphatase. The CP multilevel topography induced charge polarity and an EP and overall promoted the osteoblast phenotype of HLPSCs. The negative sign of the EP and its magnitude at the micro- and nanosockets might be sensitive factors that can trigger osteoblastic differentiation and maturation of human stromal cells.
Collapse
Affiliation(s)
- Igor A Khlusov
- Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk 634050, Russia.
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Yuri Dekhtyar
- Institute of Biomedical Engineering and Nanotechnologies, Riga Technical University, Riga LV-1658, Latvia.
| | - Yurii P Sharkeev
- Research School of High-Energy Physics, National Research Tomsk Polytechnic University, Tomsk 634050, Russia.
- Institute of Strength Physics and Materials Science of SB RAS, Tomsk 634055, Russia.
| | - Vladimir F Pichugin
- Research School of High-Energy Physics, National Research Tomsk Polytechnic University, Tomsk 634050, Russia.
| | - Marina Y Khlusova
- Department of Pathophysiology, Siberian State Medical University, Tomsk 634050, Russia.
| | - Nataliya Polyaka
- Institute of Biomedical Engineering and Nanotechnologies, Riga Technical University, Riga LV-1658, Latvia.
| | - Fedor Tyulkin
- Institute of Biomedical Engineering and Nanotechnologies, Riga Technical University, Riga LV-1658, Latvia.
| | - Viktorija Vendinya
- Institute of Biomedical Engineering and Nanotechnologies, Riga Technical University, Riga LV-1658, Latvia.
| | - Elena V Legostaeva
- Institute of Strength Physics and Materials Science of SB RAS, Tomsk 634055, Russia.
| | - Larisa S Litvinova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Valeria V Shupletsova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Olga G Khaziakhmatova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Kristina A Yurova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia.
| | - Konstantin A Prosolov
- Research School of High-Energy Physics, National Research Tomsk Polytechnic University, Tomsk 634050, Russia.
- Institute of Strength Physics and Materials Science of SB RAS, Tomsk 634055, Russia.
| |
Collapse
|
31
|
Klesen A, Jakob D, Emig R, Kohl P, Ravens U, Peyronnet R. Cardiac fibroblasts : Active players in (atrial) electrophysiology? Herzschrittmacherther Elektrophysiol 2018; 29:62-69. [PMID: 29392412 DOI: 10.1007/s00399-018-0553-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 06/07/2023]
Abstract
Fibrotic areas in cardiac muscle-be it in ventricular or atrial tissue-are considered as obstacles for conduction of the excitatory wave and can therefore facilitate re-entry, which may contribute to the sustenance of cardiac arrhythmias. Persistence of one of the most frequent arrhythmias, atrial fibrillation (AF), is accompanied by enhanced atrial fibrosis. Any kind of myocardial perturbation, whether via mechanical stress or ischemic damage, inflammation, or irregular and high-frequency electrical activity, activates fibroblasts. This leads to the secretion of paracrine factors and extracellular matrix proteins, especially collagen, and to the differentiation of fibroblasts into myofibroblasts. Excessive collagen production is the hallmark of fibrosis and impairs regular impulse propagation. In addition, direct electrical coupling between cardiomyocytes and nonmyocytes, such as fibroblasts and macrophages, via gap junctions affects conduction. Although fibroblasts are not electrically excitable, they express functional ion channels, in particular K+ channels and mechanosensitive channels, some of which could be involved in tissue remodeling. Here, we briefly review these aspects with special reference to AF.
Collapse
Affiliation(s)
- Alexander Klesen
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Dorothee Jakob
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany.
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany.
| |
Collapse
|
32
|
Ayad O, Magaud C, Sebille S, Bescond J, Mimbimi C, Cognard C, Faivre JF, Bois P, Chatelier A. Functional BKCa channel in human resident cardiac stem cells expressing W8B2. FEBS J 2017; 285:518-530. [PMID: 29211342 DOI: 10.1111/febs.14352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/02/2017] [Accepted: 11/30/2017] [Indexed: 01/24/2023]
Abstract
Recently, a new population of resident cardiac stem cells (CSCs) positive for the W8B2 marker has been identified. These CSCs are considered to be an ideal cellular source to repair myocardial damage after infarction. However, the electrophysiological profile of these cells has not been characterized yet. We first establish the conditions of isolation and expansion of W8B2+ CSCs from human heart biopsies using a magnetic sorting system followed by flow cytometry cell sorting. These cells display a spindle-shaped morphology, are highly proliferative, and possess self-renewal capacity demonstrated by their ability to form colonies. Besides, W8B2+ CSCs are positive for mesenchymal markers but negative for hematopoietic and endothelial ones. RT-qPCR and immunostaining experiments show that W8B2+ CSCs express some early cardiac-specific transcription factors but lack the expression of cardiac-specific structural genes. Using patch clamp in the whole-cell configuration, we show for the first time the electrophysiological signature of BKCa current in these cells. Accordingly, RT-PCR and western blotting analysis confirmed the presence of BKCa at both mRNA and protein levels in W8B2+ CSCs. Interestingly, BKCa channel inhibition by paxilline decreased cell proliferation in a concentration-dependent manner and halted cell cycle progression at the G0/G1 phase. The inhibition of BKCa also decreased the self-renewal capacity but did not affect migration of W8B2+ CSCs. Taken together, our results are consistent with an important role of BKCa channels in cell cycle progression and self-renewal in human cardiac stem cells.
Collapse
Affiliation(s)
- Oualid Ayad
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Christophe Magaud
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Stéphane Sebille
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Jocelyn Bescond
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Chloé Mimbimi
- Service de chirurgie cardio-thoracique, CHU Poitiers, France
| | - Christian Cognard
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Jean-Francois Faivre
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Patrick Bois
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Aurelien Chatelier
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| |
Collapse
|
33
|
Smit NW, Cócera Ortega L, Végh AMD, Meijborg VMF, Smits AM, Klerk M, Tijsen AJM, Tan HL, Goumans MJHT, Boink GJJ, Coronel R. Human Cardiomyocyte Progenitor Cells in Co-culture with Rat Cardiomyocytes Form a Pro-arrhythmic Substrate: Evidence for Two Different Arrhythmogenic Mechanisms. Front Physiol 2017; 8:797. [PMID: 29075204 PMCID: PMC5644204 DOI: 10.3389/fphys.2017.00797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/28/2017] [Indexed: 01/05/2023] Open
Abstract
Background: Cardiomyocyte progenitor cells (CMPCs) are a promising cell source for regenerative cell therapy to improve cardiac function after myocardial infarction. However, it is unknown whether undifferentiated CMPCs have arrhythmogenic risks. We investigate whether undifferentiated, regionally applied, human fetal CMPCs form a pro-arrhythmic substrate in co-culture with neonatal rat ventricular myocytes (NRVMs). Method: Unipolar extracellular electrograms, derived from micro-electrode arrays (8 × 8 electrodes) containing monolayers of NRVMs (control), or co-cultures of NRVMs and locally seeded CMPCs were used to determine conduction velocity and the incidence of tachy-arrhythmias. Micro-electrodes were used to record action potentials. Conditioned medium (Cme) of CMPCs was used to distinguish between coupling or paracrine effects. Results: Co-cultures demonstrated conduction slowing (5.6 ± 0.3 cm/s, n = 50) compared to control monolayers (13.4 ± 0.4 cm/s, n = 26) and monolayers subjected to Cme (13.7 ± 0.6 cm/s, n = 11, all p < 0.001). Furthermore, co-cultures had a more depolarized resting membrane than control monolayers (−47.3 ± 17.4 vs. −64.8 ± 7.7 mV, p < 0.001) and monolayers subjected to Cme (−64.4 ± 8.1 mV, p < 0.001). Upstroke velocity was significantly decreased in co-cultures and action potential duration was prolonged. The CMPC region was characterized by local ST-elevation in the recorded electrograms. The spontaneous rhythm was faster and tachy-arrhythmias occurred more often in co-cultured monolayers than in control monolayers (42.0 vs. 5.4%, p < 0.001). Conclusion: CMPCs form a pro-arrhythmic substrate when co-cultured with neonatal cardiomyocytes. Electrical coupling between both cell types leads to current flow between a, slowly conducting, depolarized and the normal region leading to local ST-elevations and the occurrence of tachy-arrhythmias originating from the non-depolarized zone.
Collapse
Affiliation(s)
- Nicoline W Smit
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| | - Lucia Cócera Ortega
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Anna M D Végh
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Veronique M F Meijborg
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| | - Anke M Smits
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Mischa Klerk
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Anke J M Tijsen
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hanno L Tan
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Marie-José H T Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Gerard J J Boink
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Ruben Coronel
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France
| |
Collapse
|
34
|
Hanna H, Denzi A, Liberti M, André FM, Mir LM. Electropermeabilization of Inner and Outer Cell Membranes with Microsecond Pulsed Electric Fields: Quantitative Study with Calcium Ions. Sci Rep 2017; 7:13079. [PMID: 29026094 PMCID: PMC5638809 DOI: 10.1038/s41598-017-12960-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/15/2017] [Indexed: 12/18/2022] Open
Abstract
Microsecond pulsed electric fields (μsPEF) permeabilize the plasma membrane (PM) and are widely used in research, medicine and biotechnology. For internal membranes permeabilization, nanosecond pulsed electric fields (nsPEF) are applied but this technology is complex to use. Here we report that the endoplasmic reticulum (ER) membrane can also be electropermeabilized by one 100 µs pulse without affecting the cell viability. Indeed, using Ca2+ as a permeabilization marker, we observed cytosolic Ca2+ peaks in two different cell types after one 100 µs pulse in a medium without Ca2+. Thapsigargin abolished these Ca2+ peaks demonstrating that the calcium is released from the ER. Moreover, IP3R and RyR inhibitors did not modify these peaks showing that they are due to the electropermeabilization of the ER membrane and not to ER Ca2+ channels activation. Finally, the comparison of the two cell types suggests that the PM and the ER permeabilization thresholds are affected by the sizes of the cell and the ER. In conclusion, this study demonstrates that µsPEF, which are easier to control than nsPEF, can permeabilize internal membranes. Besides, μsPEF interaction with either the PM or ER, can be an efficient tool to modulate the cytosolic calcium concentration and study Ca2+ roles in cell physiology.
Collapse
Affiliation(s)
- Hanna Hanna
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94 805, Villejuif, France
| | - Agnese Denzi
- Department of Information Engineering, Electronics and Telecommunication (DIET), University of Rome "La Sapienza", Rome, 00184, Italy
| | - Micaela Liberti
- Department of Information Engineering, Electronics and Telecommunication (DIET), University of Rome "La Sapienza", Rome, 00184, Italy
| | - Franck M André
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94 805, Villejuif, France
| | - Lluis M Mir
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94 805, Villejuif, France.
| |
Collapse
|
35
|
Choi W, Kwon SJ, Jin HJ, Jeong SY, Choi SJ, Oh W, Yang YS, Jeon HB, Jeon ES. Optimization of culture conditions for rapid clinical-scale expansion of human umbilical cord blood-derived mesenchymal stem cells. Clin Transl Med 2017; 6:38. [PMID: 29019171 PMCID: PMC5634990 DOI: 10.1186/s40169-017-0168-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have broad-spectrum therapeutic effects in various diseases, and thus have many clinical applications. However, it is difficult to produce sufficient numbers of MSCs for clinical use, and improved culture systems are required. Here, we report the effects of calcium (Ca2+) and hypoxia on the proliferation of human umbilical cord blood-derived MSCs (hUCB-MSCs). In addition, we determined the optimal conditions of these two factors for the large-scale culture of hUCB-MSCs. Methods hUCB-MSCs were maintained under hypoxic conditions (3% O2) with 1.8 mM Ca2+ during long-term culture, and their proliferation was evaluated. To characterize the underlying mechanisms, the effects on hypoxia-inducible factor (HIF)-1α and the extracellular signal-regulated kinase (ERK) signaling pathways were investigated. The therapeutic effects in a mouse emphysema model were analyzed and compared with those of naive MSCs. Results The proliferation of Ca2+/hypoxia-treated hUCB-MSCs was increased compared with that observed using either calcium or hypoxia culture alone, without loss of stem cell marker expression or differentiation ability. The enhancement of the proliferation capacity of hUCB-MSCs by the synergistic effects of Ca2+ and hypoxia was dependent on the expression of HIF-1α and the ERK signaling pathway. The proliferation of Ca2+/hypoxia-treated hUCB-MSCs resulted in a delayed senescence phenotype and increased the expression levels of stemness genes such as Oct4 and Nanog compared to those observed in conventional culture conditions. In addition, Ca2+/hypoxia-treated MSCs transplantation in the mouse emphysema model showed the same therapeutic effects as observed with naive MSCs. Conclusions These findings suggest that a Ca2+/hypoxia-based expansion system has applications for the large-scale production of MSCs for therapeutic purposes. Electronic supplementary material The online version of this article (doi:10.1186/s40169-017-0168-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wankyu Choi
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Soon-Jae Kwon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Hye Jin Jin
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Sang Young Jeong
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Soo Jin Choi
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Hong Bae Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea
| | - Eun Su Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., 21 Daewangpangyo-ro 644beon-gil, Bundang-gu, Seongnam-Si, Gyeonggi-do, 13494, Republic of Korea.
| |
Collapse
|
36
|
Sartiani L, Mannaioni G, Masi A, Novella Romanelli M, Cerbai E. The Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels: from Biophysics to Pharmacology of a Unique Family of Ion Channels. Pharmacol Rev 2017; 69:354-395. [PMID: 28878030 DOI: 10.1124/pr.117.014035] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels are important members of the voltage-gated pore loop channels family. They show unique features: they open at hyperpolarizing potential, carry a mixed Na/K current, and are regulated by cyclic nucleotides. Four different isoforms have been cloned (HCN1-4) that can assemble to form homo- or heterotetramers, characterized by different biophysical properties. These proteins are widely distributed throughout the body and involved in different physiologic processes, the most important being the generation of spontaneous electrical activity in the heart and the regulation of synaptic transmission in the brain. Their role in heart rate, neuronal pacemaking, dendritic integration, learning and memory, and visual and pain perceptions has been extensively studied; these channels have been found also in some peripheral tissues, where their functions still need to be fully elucidated. Genetic defects and altered expression of HCN channels are linked to several pathologies, which makes these proteins attractive targets for translational research; at the moment only one drug (ivabradine), which specifically blocks the hyperpolarization-activated current, is clinically available. This review discusses current knowledge about HCN channels, starting from their biophysical properties, origin, and developmental features, to (patho)physiologic role in different tissues and pharmacological modulation, ending with their present and future relevance as drug targets.
Collapse
Affiliation(s)
- Laura Sartiani
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| |
Collapse
|
37
|
Mirsadeghi S, Shahbazi E, Hemmesi K, Nemati S, Baharvand H, Mirnajafi-Zadeh J, Kiani S. Development of membrane ion channels during neural differentiation from human embryonic stem cells. Biochem Biophys Res Commun 2017; 491:166-172. [DOI: 10.1016/j.bbrc.2017.07.068] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/08/2017] [Accepted: 07/12/2017] [Indexed: 12/19/2022]
|
38
|
Recombinant human collagen-based microspheres mitigate cardiac conduction slowing induced by adipose tissue-derived stromal cells. PLoS One 2017; 12:e0183481. [PMID: 28837600 PMCID: PMC5570323 DOI: 10.1371/journal.pone.0183481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/01/2017] [Indexed: 12/22/2022] Open
Abstract
Background Stem cell therapy to improve cardiac function after myocardial infarction is hampered by poor cell retention, while it may also increase the risk of arrhythmias by providing an arrhythmogenic substrate. We previously showed that porcine adipose tissue-derived-stromal cells (pASC) induce conduction slowing through paracrine actions, whereas rat ASC (rASC) and human ASC (hASC) induce conduction slowing by direct coupling. We postulate that biomaterial microspheres mitigate the conduction slowing influence of pASC by interacting with paracrine signaling. Aim To investigate the modulation of ASC-loaded recombinant human collagen-based microspheres, on the electrophysiological behavior of neonatal rat ventricular myocytes (NRVM). Method Unipolar extracellular electrograms, derived from microelectrode arrays (8x8 electrodes) containing NRVM, co-cultured with ASC or ASC loaded microspheres, were used to determine conduction velocity (CV) and conduction heterogeneity. Conditioned medium (Cme) of (co)cultures was used to assess paracrine mechanisms. Results Microspheres did not affect CV in control (NRVM) monolayers. In co-cultures of NRVM and rASC, hASC or pASC, CV was lower than in controls (14.4±1.0, 13.0±0.6 and 9.0± 1.0 vs. 19.5±0.5 cm/s respectively, p<0.001). Microspheres loaded with either rASC or hASC still induced conduction slowing compared to controls (13.5±0.4 and 12.6±0.5 cm/s respectively, p<0.001). However, pASC loaded microspheres increased CV of NRVM compared to pASC and NRMV co-cultures (16.3±1.3 cm/s, p< 0.001) and did not differ from controls (p = NS). Cme of pASC reduced CV in control monolayers of NRVM (10.3±1.1 cm/s, p<0.001), similar to Cme derived from pASC-loaded microspheres (11.1±1.7 cm/s, p = 1.0). The presence of microspheres in monolayers of NRVM abolished the CV slowing influence of Cme pASC (15.9±1.0 cm/s, p = NS vs. control). Conclusion The application of recombinant human collagen-based microspheres mitigates indirect paracrine conduction slowing through interference with a secondary autocrine myocardial factor.
Collapse
|
39
|
Petecchia L, Usai C, Vassalli M, Gavazzo P. Biophysical characterization of nanostructured TiO 2 as a good substrate for hBM-MSC adhesion, growth and differentiation. Exp Cell Res 2017; 358:111-119. [PMID: 28619648 DOI: 10.1016/j.yexcr.2017.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/10/2017] [Accepted: 06/08/2017] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells from human bone marrow (hBM-MSC) are widely utilized for clinical applications involving bone healing. In this context, their use has been often optimized in association to variously designed titanium substrates, being this material of great use in orthopaedic implants. According to recent findings, the ability of hBM-MSC to differentiate towards a specific lineage is not only driven by biochemical signals, but physical stimuli, such as rigidity or roughness of the substrate, can also support a commitment towards osteogenic differentiation. Moreover, the presence of features with defined dimensional scales, in particular nanometer-size, also proved to elicit specific biological effects. Here we evaluated the effectiveness of a nano-patterned titanium surface in sustaining hBM-MSC adhesion, growth and differentiation by means of a panel of biophysical tools: morphometry, electrophysiology, intracellular calcium measurements and immunocytochemistry. The results substantiate the idea that this micro-textured titanium dioxide is a good surface for growth and differentiation of hBM-MSC and it exhibits a stimulating action mainly in the initial period of differentiation. Moreover, the basal concentration of free cytosolic Calcium [Ca2+]i is confirmed to be a good hallmark of the hBM-MSC maturation stage. The study could provide relevant hints to help improving the biocompatibility and osteointegration potential of clinical titanium implants.
Collapse
Affiliation(s)
- L Petecchia
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genova, Italy
| | - C Usai
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genova, Italy
| | - M Vassalli
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genova, Italy
| | - P Gavazzo
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genova, Italy.
| |
Collapse
|
40
|
Kim E, Park WS, Hong SH. Expression of Ion Channels in Perivascular Stem Cells derived from Human Umbilical Cords. Dev Reprod 2017; 21:11-18. [PMID: 28484740 PMCID: PMC5409206 DOI: 10.12717/dr.2017.21.1.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 12/29/2016] [Accepted: 01/05/2017] [Indexed: 11/17/2022]
Abstract
Potassium channels, the largest group of pore proteins, selectively regulate the flow of potassium (K+) ions across cell membranes. The activity and expression of K+ channels are critical for the maintenance of normal functions in vessels and neurons, and for the regulation of cell differentiation and maturation. However, their role and expression in stem cells have been poorly understood. In this study, we isolated perivascular stem cells (PVCs) from human umbilical cords and investigated the expression patterns of big-conductance Ca2+-activated K+ (BKCa) and voltage-dependent K+ (Kv) channels using the reverse transcription polymerase chain reaction. We also examined the effect of high glucose (HG, 25 mM) on expression levels of BKCa and Kv channels in PVCs. KCa1.1, KCaβ3, Kv1.3, Kv3.2, and Kv6.1 were detected in undifferentiated PVCs. In addition, HG treatment increased the amounts of BKCaβ3a, BKCaβ4, Kv1.3, Kv1.6, and Kv6.1 transcripts. These results suggested that ion channels may have important functions in the growth and differentiation of PVCs, which could be influenced by HG exposure.
Collapse
Affiliation(s)
- Eunbi Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
41
|
Calcium-gated K+ channels of the KCa1.1- and KCa3.1-type couple intracellular Ca2+ signals to membrane hyperpolarization in mesenchymal stromal cells from the human adipose tissue. Pflugers Arch 2016; 469:349-362. [DOI: 10.1007/s00424-016-1932-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/10/2016] [Accepted: 12/14/2016] [Indexed: 01/06/2023]
|
42
|
Wang M, Hu Y, Fan Y, Guo Y, Chen F, Chen S, Li Q, Chen Z. Involvement of Hydrogen Sulfide in Endothelium-Derived Relaxing Factor-Mediated Responses in Rat Cerebral Arteries. J Vasc Res 2016; 53:172-185. [DOI: 10.1159/000448712] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/26/2016] [Indexed: 11/19/2022] Open
|
43
|
Thi Do D, Phan NN, Wang CY, Sun Z, Lin YC. Novel regulations of MEF2-A, MEF2-D, and CACNA1S in the functional incompetence of adipose-derived mesenchymal stem cells by induced indoxyl sulfate in chronic kidney disease. Cytotechnology 2016; 68:2589-2604. [PMID: 27550174 DOI: 10.1007/s10616-016-9983-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 05/12/2016] [Indexed: 12/13/2022] Open
Abstract
Indoxyl sulfate (IS) is a digestive intermediate product that is a known indicator of chronic kidney disease. Its toxicity has also been suggested to accelerate chronic kidney disease. Recently, mesenchymal stem cells (MSCs) have been confirmed as a potential treatment in kidney regeneration. To determine the universal alteration in gene expression, we combined high-throughput microarray technology and in vitro culture of adipose-derived mesenchymal stem cells at different doses of IS (20, 40, 60 mg/l). We found that indoxyl sulfate has a remarkable interconnection with stem cell and calcium/calmodulin-dependent kinase pathways. In vitro results showed that indoxyl sulfate exerts anti-proliferation and anti-migration effects on ADMSCs. In addition, IS effects lead to increase in apoptotic cells and cells arrested at the G1 phase. Moreover, MEF2-A, MEF2-D and CACNA1S expression significantly decreased after indoxyl sulfate treatment. It can be speculated that following treatment with indoxyl sulfate, the function of ADMSCs is decreased and ADMSCs' ability to support renal tubule regeneration in chronic kidney disease patients may be lower.
Collapse
Affiliation(s)
- Duyen Thi Do
- Faculty of Applied Sciences, Ton Duc Thang University, Nguyen Huu Tho St., Tan Phong Ward, Dist. 7, Ho Chi Minh City, Vietnam.,Graduate Institute of Biotechnology, Chinese Culture University, 55. Hwa-Kang Rd., Yang-Ming-Shan, Taipei, 11114, Taiwan
| | - Nam Nhut Phan
- Faculty of Applied Sciences, Ton Duc Thang University, Nguyen Huu Tho St., Tan Phong Ward, Dist. 7, Ho Chi Minh City, Vietnam
| | - Chih-Yang Wang
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Zhengda Sun
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Yen-Chang Lin
- Graduate Institute of Biotechnology, Chinese Culture University, 55. Hwa-Kang Rd., Yang-Ming-Shan, Taipei, 11114, Taiwan.
| |
Collapse
|
44
|
ten Sande JN, Smit NW, Parvizi M, van Amersfoorth SC, Plantinga JA, van Dessel PF, de Bakker JM, Harmsen MC, Coronel R. Differential Mechanisms of Myocardial Conduction Slowing by Adipose Tissue-Derived Stromal Cells Derived from Different Species. Stem Cells Transl Med 2016; 6:22-30. [PMID: 28170198 PMCID: PMC5442737 DOI: 10.5966/sctm.2015-0415] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/22/2016] [Indexed: 11/18/2022] Open
Abstract
Stem cell therapy is a promising therapeutic option to treat patients after myocardial infarction. However, the intramyocardial administration of large amounts of stem cells might generate a proarrhythmic substrate. Proarrhythmic effects can be explained by electrotonic and/or paracrine mechanisms. The narrow therapeutic time window for cell therapy and the presence of comorbidities limit the application of autologous cell therapy. The use of allogeneic or xenogeneic stem cells is a potential alternative to autologous cells, but differences in the proarrhythmic effects of adipose‐derived stromal cells (ADSCs) across species are unknown. Using microelectrode arrays and microelectrode recordings, we obtained local unipolar electrograms and action potentials from monolayers of neonatal rat ventricular myocytes (NRVMs) that were cocultured with rat, human, or pig ADSCs (rADSCs, hADSCs, pADSCs, respectively). Monolayers of NRVMs were cultured in the respective conditioned medium to investigate paracrine effects. We observed significant conduction slowing in all cardiomyocyte cultures containing ADSCs, independent of species used (p < .01). All cocultures were depolarized compared with controls (p < .01). Only conditioned medium taken from cocultures with pADSCs and applied to NRVM monolayers demonstrated similar electrophysiological changes as the corresponding cocultures. We have shown that independent of species used, ADSCs cause conduction slowing in monolayers of NRVMs. In addition, pADSCs exert conduction slowing mainly by a paracrine effect, whereas the influence on conduction by hADSCs and rADSCs is preferentially by electrotonic interaction. Stem Cells Translational Medicine2017;6:22–30
Collapse
Affiliation(s)
- Judith N. ten Sande
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Netherlands Heart Institute, Utrecht, The Netherlands
| | - Nicoline W. Smit
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Netherlands Heart Institute, Utrecht, The Netherlands
| | - Mojtaba Parvizi
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Shirley C.M. van Amersfoorth
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Josée A. Plantinga
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Pascal F.H.M. van Dessel
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jacques M.T. de Bakker
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Netherlands Heart Institute, Utrecht, The Netherlands
| | - Marco C. Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Ruben Coronel
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- L'Institut de Rythmologie et de Modélisation Cardiaque, Université Bordeaux, Segalen, Bordeaux, France
| |
Collapse
|
45
|
Mayourian J, Savizky RM, Sobie EA, Costa KD. Modeling Electrophysiological Coupling and Fusion between Human Mesenchymal Stem Cells and Cardiomyocytes. PLoS Comput Biol 2016; 12:e1005014. [PMID: 27454812 PMCID: PMC4959759 DOI: 10.1371/journal.pcbi.1005014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 06/08/2016] [Indexed: 01/16/2023] Open
Abstract
Human mesenchymal stem cell (hMSC) delivery has demonstrated promise in preclinical and clinical trials for myocardial infarction therapy; however, broad acceptance is hindered by limited understanding of hMSC-human cardiomyocyte (hCM) interactions. To better understand the electrophysiological consequences of direct heterocellular connections between hMSCs and hCMs, three original mathematical models were developed, representing an experimentally verified triad of hMSC families with distinct functional ion channel currents. The arrhythmogenic risk of such direct electrical interactions in the setting of healthy adult myocardium was predicted by coupling and fusing these hMSC models to the published ten Tusscher midcardial hCM model. Substantial variations in action potential waveform—such as decreased action potential duration (APD) and plateau height—were found when hCMs were coupled to the two hMSC models expressing functional delayed rectifier-like human ether à-go-go K+ channel 1 (hEAG1); the effects were exacerbated for fused hMSC-hCM hybrid cells. The third family of hMSCs (Type C), absent of hEAG1 activity, led to smaller single-cell action potential alterations during coupling and fusion, translating to longer tissue-level mean action potential wavelength. In a simulated 2-D monolayer of cardiac tissue, re-entry vulnerability with low (5%) hMSC insertion was approximately eight-fold lower with Type C hMSCs compared to hEAG1-functional hMSCs. A 20% decrease in APD dispersion by Type C hMSCs compared to hEAG1-active hMSCs supports the claim of reduced arrhythmogenic potential of this cell type with low hMSC insertion. However, at moderate (15%) and high (25%) hMSC insertion, the vulnerable window increased independent of hMSC type. In summary, this study provides novel electrophysiological models of hMSCs, predicts possible arrhythmogenic effects of hMSCs when directly coupled to healthy hCMs, and proposes that isolating a subset of hMSCs absent of hEAG1 activity may offer increased safety as a cell delivery cardiotherapy at low levels of hMSC-hCM coupling. Myocardial infarction—better known as a heart attack—strikes on average every 43 seconds in America. An emerging approach to treat myocardial infarction patients involves the delivery of human mesenchymal stem cells (hMSCs) to the damaged heart. While clinical trials of this therapeutic approach have yet to report adverse effects on heart electrical rhythm, such consequences have been implicated in simpler experimental systems and thus remain a concern. In this study, we utilized mathematical modeling to simulate electrical interactions arising from direct coupling between hMSCs and human heart cells to develop insight into the possible adverse effects of this therapeutic approach on human heart electrical activity, and to assess a novel strategy for reducing some potential risks of this therapy. We developed the first mathematical models of electrical activity of three families of hMSCs based on published experimental data, and integrated these with previously established mathematical models of human heart cell electrical activity. Our computer simulations demonstrated that one particular family of hMSCs minimized the disturbances in cardiac electrical activity both at the single-cell and tissue levels, suggesting that isolating this specific sub-population of hMSCs for myocardial delivery could potentially increase the safety of future hMSC-based heart therapies.
Collapse
Affiliation(s)
- Joshua Mayourian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ruben M. Savizky
- Department of Chemistry, The Cooper Union, New York, New York, United States of America
| | - Eric A. Sobie
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kevin D. Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
46
|
Squecco R, Idrizaj E, Morelli A, Gallina P, Vannelli GB, Francini F. An electrophysiological study on the effects of BDNF and FGF2 on voltage dependent Ca(2+) currents in developing human striatal primordium. Mol Cell Neurosci 2016; 75:50-62. [PMID: 27370937 DOI: 10.1016/j.mcn.2016.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/24/2016] [Accepted: 06/27/2016] [Indexed: 01/06/2023] Open
Abstract
Over the past decades, studies in both Huntington's disease animal models and pilot clinical trials have demonstrated that replacement of degenerated striatum and repair of circuitries by grafting fetal striatal primordium is feasible, safe and may counteract disease progression. However, a better comprehension of striatal ontogenesis is required to assess the fetal graft regenerative potential. During neuronal development, neurotrophins exert pleiotropic actions in regulating cell fate and synaptic plasticity. In this regard, brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF2) are crucially implicated in the control of fate choice of striatal progenitor cells. In this study, we intended to refine the functional features of human striatal precursor (HSP) cells isolated from ganglionic eminence of 9-12week old human fetuses, by studying with electrophysiological methods the effect of BDNF and FGF2 on the membrane biophysical properties and the voltage-dependent Ca(2+) currents. These features are particularly relevant to evaluate neuronal cell functioning and can be considered reliable markers of the developmental phenotype of human striatal primordium. Our results have demonstrated that BDNF and FGF2 induced membrane hyperpolarization, increased the membrane capacitance and reduced the resting total and specific conductance values, suggesting a more efficient control of resting ionic fluxes. Moreover, the treatment with both neurotrophins enhanced N-type Ca(2+) current amplitude and reduced L- and T-type ones. Overall, our data indicate that BDNF and FGF2 may help HSP cells to attain a more functionally mature phenotype.
Collapse
Affiliation(s)
- Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, viale Morgagni 63, 50134 Florence, Italy.
| | - Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, viale Morgagni 63, 50134 Florence, Italy
| | - Annamaria Morelli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Pasquale Gallina
- Department of Surgery and Translational Medicine, University of Florence, Largo Palagi 1, 50139 Florence, Italy
| | - Gabriella B Vannelli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Fabio Francini
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, viale Morgagni 63, 50134 Florence, Italy
| |
Collapse
|
47
|
Expression of KCNA5 Protein in Human Mammary Epithelial Cell Line Associated with Caveolin-1. J Membr Biol 2016; 249:449-57. [DOI: 10.1007/s00232-016-9885-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/06/2016] [Indexed: 11/24/2022]
|
48
|
Forostyak O, Butenko O, Anderova M, Forostyak S, Sykova E, Verkhratsky A, Dayanithi G. Specific profiles of ion channels and ionotropic receptors define adipose- and bone marrow derived stromal cells. Stem Cell Res 2016; 16:622-34. [PMID: 27062357 DOI: 10.1016/j.scr.2016.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 01/09/2023] Open
Abstract
Adherent, fibroblastic cells from different tissues are thought to contain subsets of tissue-specific stem/progenitor cells (often called mesenchymal stem cells). These cells display similar cell surface characteristics based on their fibroblastic nature, but also exhibit differences in molecular phenotype, growth rate, and their ability to differentiate into various cell phenotypes. The mechanisms underlying these differences remain poorly understood. We analyzed Ca(2+) signals and membrane properties in rat adipose-derived stromal cells (ADSCs) and bone marrow stromal cells (BMSCs) in basal conditions, and then following a switch into medium that contains factors known to modify their character. Modified ADSCs (mADSCs) expressed L-type Ca(2+) channels whereas both L- and P/Q- channels were operational in mBMSCs. Both mADSCs and mBMSCs possessed functional endoplasmic reticulum Ca(2+) stores, expressed ryanodine receptor-1 and -3, and exhibited spontaneous [Ca(2+)]i oscillations. The mBMSCs expressed P2X7 purinoceptors; the mADSCs expressed both P2X (but not P2X7) and P2Y (but not P2Y1) receptors. Both types of stromal cells exhibited [Ca(2+)]i responses to vasopressin (AVP) and expressed V1 type receptors. Functional oxytocin (OT) receptors were, in contrast, expressed only in modified ADSCs and BMSCs. AVP and OT-induced [Ca(2+)]i responses were dose-dependent and were blocked by their respective specific receptor antagonists. Electrophysiological data revealed that passive ion currents dominated the membrane conductance in ADSCs and BMSCs. Medium modification led to a significant shift in the reversal potential of passive currents from -40 to -50mV in cells in basal to -80mV in modified cells. Hence membrane conductance was mediated by non-selective channels in cells in basal conditions, whereas in modified medium conditions, it was associated with K(+)-selective channels. Our results indicate that modification of ADSCs and BMSCs by alteration in medium formulation is associated with significant changes in their Ca(2+) signaling and membrane properties.
Collapse
Affiliation(s)
- Oksana Forostyak
- Department of Molecular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic; Department of Neuroscience, Charles University, Second Faculty of Medicine, V Uvalu 84, Prague 15006, Czech Republic
| | - Olena Butenko
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic.
| | - Miroslava Anderova
- Department of Neuroscience, Charles University, Second Faculty of Medicine, V Uvalu 84, Prague 15006, Czech Republic; Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Serhiy Forostyak
- Department of Neuroscience, Charles University, Second Faculty of Medicine, V Uvalu 84, Prague 15006, Czech Republic; Department of Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Eva Sykova
- Department of Neuroscience, Charles University, Second Faculty of Medicine, V Uvalu 84, Prague 15006, Czech Republic; Department of Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Alexei Verkhratsky
- University of Manchester, School of Biological Sciences, D.4417 Michael Smith Building, Oxford Road, Manchester M13 9PT, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain; University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Govindan Dayanithi
- Department of Molecular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic; Institut National de la Santé et de la Recherche Médicale-U1198, Université Montpellier, Montpellier 34095, France; Ecole Pratique des Hautes Etudes-Sorbonne, Les Patios Saint-Jacques, 4-14 rue Ferrus, 75014 Paris, France.
| |
Collapse
|
49
|
Forostyak O, Forostyak S, Kortus S, Sykova E, Verkhratsky A, Dayanithi G. Physiology of Ca2+ signalling in stem cells of different origins and differentiation stages. Cell Calcium 2016; 59:57-66. [DOI: 10.1016/j.ceca.2016.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 01/28/2016] [Accepted: 02/02/2016] [Indexed: 01/15/2023]
|
50
|
Grajales L, Lach LE, Janisch P, Geenen DL, García J. Temporal expression of calcium channel subunits in satellite cells and bone marrow mesenchymal cells. Stem Cell Rev Rep 2016; 11:408-22. [PMID: 25277766 DOI: 10.1007/s12015-014-9566-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSC) can be differentiated into myocytes, as well as adipocytes, chondrocytes, and osteocytes in culture. Calcium channels mediate excitation-contraction coupling and are essential for the function of muscle. However, little is known about the expression of calcium channel subunits and calcium handling in stem cells. We examined whether the expression of calcium channel subunits in MSC is similar to that of skeletal muscle satellite cells and if their levels of expression are modified after treatment with bone morphogenetic protein-4 (BMP4). We found that during myogenic differentiation, MSC first express the α2δ1 subunit and the cardiac channel subunit Cav1.2. In contrast to the α2δ1 subunit levels, the Cav1.2 subunit decreases rapidly with time. The skeletal channel subunit Cav1.1 is detected at day 3 but its expression increases considerably, resembling more closely the expression of the subunits in satellite cells. Treatment of MSC with BMP4 caused a significant increase in expression of Cav1.2, a delay in expression of Cav1.1, and a reduction in the duration of calcium transients when extracellular calcium was removed. Calcium currents and transients followed a pattern related to the expression of the cardiac (Cav1.2) or skeletal (Cav1.1) α1subunits. These results indicate that differentiation of untreated MSC resembles differentiation of skeletal muscle and that BMP4 reduces skeletal muscle calcium channel expression and promotes the expression of cardiac calcium channels during myogenic differentiation.
Collapse
Affiliation(s)
- Liliana Grajales
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Ave, Chicago, IL, 60612, USA
| | | | | | | | | |
Collapse
|