1
|
Liu X, Zhang M, Wang P, Zheng K, Wang X, Xie W, Pan X, Shen R, Liu R, Ding J, Wei Q. Nanoscale distribution of bioactive ligands on biomaterials regulates cell mechanosensing through translocation of actin into the nucleus. Proc Natl Acad Sci U S A 2025; 122:e2501264122. [PMID: 40042901 PMCID: PMC11912452 DOI: 10.1073/pnas.2501264122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Cells respond to adhesive ligands such as arginine-glycine-aspartate (RGD) through integrins, which regulates cellular activities via influencing cytoskeleton assembly. Herein, we report that the nanoscale distribution of active ligands on biomaterials regulates cells through not only cytoplasmic tension but also nuclear tension. This is particularly related to translocation of actin into nucleus and highlighted in our interpretation of an "abnormal" phenomenon that large RGD nanospacing (>70 nm) disassembles integrin clusters, inhibits cell adhesion, but promotes osteogenic differentiation of mesenchymal stem cells. Our studies reveal that the unstable adhesion at the 150 nm RGD distance increases actin dynamics, resulting in the nuclear translocation of globular (G) actin. The compartment polymerization of more G-actins to filamentous actins in nucleus increases nuclear tension, facilitating transcription activity and releasing calcium ions from the endoplasmic reticulum. This noncanonical mechanotransduction process sheds insight into mechanotransduction pertinent to cell-material interactions.
Collapse
Affiliation(s)
- Xiaojing Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan250012, China
| | - Man Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Biomedical Engineering, Sichuan University, Chengdu610065, China
| | - Peng Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
- Sauvage Laboratory for Smart Materials, School of Integrated Circuits, Harbin Institute of Technology, Shenzhen518055, China
| | - Kaikai Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| | - Xinlei Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Wenyan Xie
- National Key Laboratory of Biotherapy, Sichuan University, Chengdu610041, China
| | - Xiaokai Pan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| | - Runjia Shen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Qiang Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| |
Collapse
|
2
|
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
|
3
|
Jeung SY, An JH, Kim SS, Youn HY. Safety of Gonadal Tissue-Derived Mesenchymal Stem Cell Therapy in Geriatric Dogs with Chronic Disease. Animals (Basel) 2024; 14:2134. [PMID: 39061596 PMCID: PMC11273526 DOI: 10.3390/ani14142134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Ensuring the safety of mesenchymal stem cell (MSC) therapy is a fundamental requirement in clinical practice. This study aimed to assess the safety of using gonadal tissue-derived MSCs (n = 10) compared to the commonly utilized adipose tissue-derived MSCs (n = 9) in geriatric dogs with chronic diseases. All participants received allogeneic MSC therapy, and no allergic reactions due to allogeneic cell immunogenicity were noted. Both groups showed no adverse changes in physical exams or hematological parameters before and after therapy. Importantly, there were no instances of tumor formation or growth post-treatment in either group. The findings demonstrated that dogs treated with gonadal tissue-derived MSCs experienced no clinical adverse effects. However, clinical adverse effects were reported in one case of adipose tissue-derived MSC therapy. Despite limitations in monitoring beyond one year and constraints due to a small and diverse patient group, this pioneering study validates the safe use of gonadal tissue-derived MSCs in aged companion animals. It underscores the potential of utilizing tissues from neutering procedures to advance regenerative medicine and expand cell banks and therapy options for companion animals.
Collapse
Affiliation(s)
- So-Young Jeung
- VIP Animal Medical Center, Seoul 02830, Republic of Korea; (S.-Y.J.); (S.-S.K.)
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ju-Hyun An
- Laboratory of Veterinary Emergency and Critical Care, Department of Veterinary Clinical Science, College of Veterinary Medicine, Kangwon National University, Chuncheon-si 24341, Republic of Korea;
| | - Sung-Soo Kim
- VIP Animal Medical Center, Seoul 02830, Republic of Korea; (S.-Y.J.); (S.-S.K.)
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
4
|
Qin S, Niu Y, Zhang Y, Wang W, Zhou J, Bai Y, Ma G. Metal Ion-Containing Hydrogels: Synthesis, Properties, and Applications in Bone Tissue Engineering. Biomacromolecules 2024; 25:3217-3248. [PMID: 38237033 DOI: 10.1021/acs.biomac.3c01072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Hydrogel, as a unique scaffold material, features a three-dimensional network system that provides conducive conditions for the growth of cells and tissues in bone tissue engineering (BTE). In recent years, it has been discovered that metal ion-containing hybridized hydrogels, synthesized with metal particles as the foundation, exhibit excellent physicochemical properties, osteoinductivity, and osteogenic potential. They offer a wide range of research prospects in the field of BTE. This review provides an overview of the current state and recent advancements in research concerning metal ion-containing hydrogels in the field of BTE. Within materials science, it covers topics such as the binding mechanisms of metal ions within hydrogel networks, the types and fabrication methods of various metal ion-containing hydrogels, and the influence of metal ions on the properties of hydrogels. In the context of BTE, the review delves into the osteogenic mechanisms of various metal ions, the applications of metal ion-containing hydrogels in BTE, and relevant experimental studies in vitro and in vivo. Furthermore, future improvements in bone repair can be anticipated through advancements in bone bionics, exploring interactions between metal ions and the development of a wider range of metal ions and hydrogel types.
Collapse
Affiliation(s)
- Shengao Qin
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, P. R. China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, P. R. China
| | - Yimeng Niu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, P. R. China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, P. R. China
| | - Yihan Zhang
- School of Stomatology, Harbin Medical University, Harbin 150020, P. R. China
| | - Weiyi Wang
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, P. R. China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, P. R. China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, P. R. China
- Department of VIP Dental Service, School of Stomatology, Capital Medical University, Beijing 100050, P. R. China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P. R. China
| | - Yingjie Bai
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, P. R. China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, P. R. China
| | - Guowu Ma
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshunnan Road, Dalian 116044, P. R. China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Lvshun South Road, Dalian 116044, P. R. China
- Department of Stomatology, Stomatological Hospital Affiliated School of Stomatology of Dalian Medical University, No. 397 Huangpu Road, Shahekou District, Dalian 116086, P. R. China
| |
Collapse
|
5
|
Wu S, Zhou H, Ling H, Sun Y, Luo Z, Ngo T, Fu Y, Wang W, Kong Y. LIPUS regulates the progression of knee osteoarthritis in mice through primary cilia-mediated TRPV4 channels. Apoptosis 2024; 29:785-798. [PMID: 38517601 PMCID: PMC11055729 DOI: 10.1007/s10495-024-01950-9] [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] [Accepted: 02/24/2024] [Indexed: 03/24/2024]
Abstract
Osteoarthritis (OA) is a common disease in middle-aged and elderly people. An imbalance in calcium ion homeostasis will contribute to chondrocyte apoptosis and ultimately lead to the progression of OA. Transient receptor potential channel 4 (TRPV4) is involved in the regulation of intracellular calcium homeostasis. TRPV4 is expressed in primary cilia, which can sense mechanical stimuli from outside the cell, and its abnormal expression is closely related to the development of OA. Low-intensity pulsed ultrasound (LIPUS) can alleviate chondrocyte apoptosis while the exact mechanism is unclear. In this project, with the aim of revealing the mechanism of action of LIPUS, we proposed to use OA chondrocytes and animal models, LIPUS intervention, inhibition of primary cilia, use TRPV4 inhibitors or TRPV4 agonist, and use Immunofluorescence (IF), Immunohistochemistry (IHC), Western Blot (WB), Quantitative Real-time PCR (QP) to detect the expression of cartilage synthetic matrix and endoplasmic reticulum stress markers. The results revealed that LIPUS altered primary cilia expression, promoted synthetic matrix metabolism in articular chondrocytes and was associated with primary cilia. In addition, LIPUS exerted a active effect on OA by activating TRPV4, inducing calcium inward flow, and facilitating the entry of NF-κB into the nucleus to regulate synthetic matrix gene transcription. Inhibition of TRPV4 altered primary cilia expression in response to LIPUS stimulation, and knockdown of primary cilia similarly inhibited TRPV4 function. These results suggest that LIPUS mediates TRPV4 channels through primary cilia to regulate the process of knee osteoarthritis in mice.
Collapse
Affiliation(s)
- Sha Wu
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haiqi Zhou
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huixian Ling
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuyan Sun
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ziyu Luo
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - ThaiNamanh Ngo
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuanyuan Fu
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wen Wang
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Kong
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
6
|
Uzieliene I, Popov A, Vaiciuleviciute R, Kirdaite G, Bernotiene E, Ramanaviciene A. Polypyrrole-based structures for activation of cellular functions under electrical stimulation. Bioelectrochemistry 2024; 155:108585. [PMID: 37847982 DOI: 10.1016/j.bioelechem.2023.108585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/04/2023] [Accepted: 10/08/2023] [Indexed: 10/19/2023]
Abstract
Polypyrrole (Ppy) is an electroconductive polymer used in various applications, including in vitro experiments with cell cultures under electrical stimulation (ES). Ppy can be applied in various forms and most importantly, it is biocompatible with cells. Ppy specifically directs ES to cells, which makes Ppy a potential polymer for the development of novel technologies for targeted tissue regeneration. The high potential of ES in combination with different Ppy-based systems, such as hydrogels, scaffolds, or Ppy-layers is advantageous to stimulate cellular differentiation towards neurogenic, cardiac, muscle, and osteogenic lineages. Different in-house devices and the principles of ES application used to stimulate cellular functions are reviewed and summarized. The focus of this review is to observe the most relevant studies and their in-house techniques regarding the application of Ppy-based materials for the use of bone, neural, cardiac, and muscle tissue regeneration under ES. Different types of Ppy materials, such as Ppy particles, layers/films, membranes, and 3D-shaped synthetic and natural scaffolds, as well as combining Ppy with different active molecules are reviewed.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Anton Popov
- Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; NanoTechnas - Center on Nanotechnology and Materials Sciences, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko g. 24, LT-03225 Vilnius, Lithuania
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Gailute Kirdaite
- Department of Experimental, Preventive and Clinical Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; Faculty of Fundamental Sciences, Vilnius Gediminas Technical University, VilniusTech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Almira Ramanaviciene
- Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; NanoTechnas - Center on Nanotechnology and Materials Sciences, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko g. 24, LT-03225 Vilnius, Lithuania.
| |
Collapse
|
7
|
George LF, Follmer ML, Fontenoy E, Moran HR, Brown JR, Ozekin YH, Bates EA. Endoplasmic Reticulum Calcium Mediates Drosophila Wing Development. Bioelectricity 2023; 5:290-306. [PMID: 38143873 PMCID: PMC10733776 DOI: 10.1089/bioe.2022.0036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023] Open
Abstract
Background The temporal dynamics of morphogen presentation impacts transcriptional responses and tissue patterning. However, the mechanisms controlling morphogen release are far from clear. We found that inwardly rectifying potassium (Irk) channels regulate endogenous transient increases in intracellular calcium and bone morphogenetic protein (BMP/Dpp) release for Drosophila wing development. Inhibition of Irk channels reduces BMP/Dpp signaling, and ultimately disrupts wing morphology. Ion channels impact development of several tissues and organisms in which BMP signaling is essential. In neurons and pancreatic beta cells, Irk channels modulate membrane potential to affect intracellular Ca++ to control secretion of neurotransmitters and insulin. Based on Irk activity in neurons, we hypothesized that electrical activity controls endoplasmic reticulum (ER) Ca++ release into the cytoplasm to regulate the release of BMP. Materials and Methods To test this hypothesis, we reduced expression of four proteins that control ER calcium, Stromal interaction molecule 1 (Stim), Calcium release-activated calcium channel protein 1 (Orai), SarcoEndoplasmic Reticulum Calcium ATPase (SERCA), small conductance calcium-activated potassium channel (SK), and Bestrophin 2 (Best2) using RNAi and documented wing phenotypes. We use live imaging to study calcium and Dpp release within pupal wings and larval wing discs. Additionally, we employed immunohistochemistry to characterize Small Mothers Against Decapentaplegic (SMAD) phosphorylation downstream of the BMP/Dpp pathway following RNAi knockdown. Results We found that reduced Stim and SERCA function decreases amplitude and frequency of endogenous calcium transients in the wing disc and reduced BMP/Dpp release. Conclusion Our results suggest control of ER calcium homeostasis is required for BMP/Dpp release, and Drosophila wing development.
Collapse
Affiliation(s)
- Laura Faith George
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mikaela Lynn Follmer
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily Fontenoy
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Hannah Rose Moran
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jeremy Ryan Brown
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yunus H. Ozekin
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily Anne Bates
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
8
|
Moccia F, Brunetti V, Soda T, Faris P, Scarpellino G, Berra-Romani R. Store-Operated Ca 2+ Entry as a Putative Target of Flecainide for the Treatment of Arrhythmogenic Cardiomyopathy. J Clin Med 2023; 12:5295. [PMID: 37629337 PMCID: PMC10455538 DOI: 10.3390/jcm12165295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/04/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic disorder that may lead patients to sudden cell death through the occurrence of ventricular arrhythmias. ACM is characterised by the progressive substitution of cardiomyocytes with fibrofatty scar tissue that predisposes the heart to life-threatening arrhythmic events. Cardiac mesenchymal stromal cells (C-MSCs) contribute to the ACM by differentiating into fibroblasts and adipocytes, thereby supporting aberrant remodelling of the cardiac structure. Flecainide is an Ic antiarrhythmic drug that can be administered in combination with β-adrenergic blockers to treat ACM due to its ability to target both Nav1.5 and type 2 ryanodine receptors (RyR2). However, a recent study showed that flecainide may also prevent fibro-adipogenic differentiation by inhibiting store-operated Ca2+ entry (SOCE) and thereby suppressing spontaneous Ca2+ oscillations in C-MSCs isolated from human ACM patients (ACM C-hMSCs). Herein, we briefly survey ACM pathogenesis and therapies and then recapitulate the main molecular mechanisms targeted by flecainide to mitigate arrhythmic events, including Nav1.5 and RyR2. Subsequently, we describe the role of spontaneous Ca2+ oscillations in determining MSC fate. Next, we discuss recent work showing that spontaneous Ca2+ oscillations in ACM C-hMSCs are accelerated to stimulate their fibro-adipogenic differentiation. Finally, we describe the evidence that flecainide suppresses spontaneous Ca2+ oscillations and fibro-adipogenic differentiation in ACM C-hMSCs by inhibiting constitutive SOCE.
Collapse
Affiliation(s)
- Francesco Moccia
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.B.); (G.S.)
| | - Valentina Brunetti
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.B.); (G.S.)
| | - Teresa Soda
- Department of Health Sciences, University of Magna Graecia, 88100 Catanzaro, Italy;
| | - Pawan Faris
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Giorgia Scarpellino
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.B.); (G.S.)
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla 72410, Mexico;
| |
Collapse
|
9
|
Takács R, Kovács P, Ebeid RA, Almássy J, Fodor J, Ducza L, Barrett-Jolley R, Lewis R, Matta C. Ca2+-Activated K+ Channels in Progenitor Cells of Musculoskeletal Tissues: A Narrative Review. Int J Mol Sci 2023; 24:ijms24076796. [PMID: 37047767 PMCID: PMC10095002 DOI: 10.3390/ijms24076796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Musculoskeletal disorders represent one of the main causes of disability worldwide, and their prevalence is predicted to increase in the coming decades. Stem cell therapy may be a promising option for the treatment of some of the musculoskeletal diseases. Although significant progress has been made in musculoskeletal stem cell research, osteoarthritis, the most-common musculoskeletal disorder, still lacks curative treatment. To fine-tune stem-cell-based therapy, it is necessary to focus on the underlying biological mechanisms. Ion channels and the bioelectric signals they generate control the proliferation, differentiation, and migration of musculoskeletal progenitor cells. Calcium- and voltage-activated potassium (KCa) channels are key players in cell physiology in cells of the musculoskeletal system. This review article focused on the big conductance (BK) KCa channels. The regulatory function of BK channels requires interactions with diverse sets of proteins that have different functions in tissue-resident stem cells. In this narrative review article, we discuss the main ion channels of musculoskeletal stem cells, with a focus on calcium-dependent potassium channels, especially on the large conductance BK channel. We review their expression and function in progenitor cell proliferation, differentiation, and migration and highlight gaps in current knowledge on their involvement in musculoskeletal diseases.
Collapse
Affiliation(s)
- Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Rana Abdelsattar Ebeid
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, Semmelweis University, H-1428 Budapest, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Ducza
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 3GA, UK
| | - Rebecca Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
10
|
Uzieliene I, Bironaite D, Miksiunas R, Bagdonas E, Vaiciuleviciute R, Mobasheri A, Bernotiene E. The Effect of CaV1.2 Inhibitor Nifedipine on Chondrogenic Differentiation of Human Bone Marrow or Menstrual Blood-Derived Mesenchymal Stem Cells and Chondrocytes. Int J Mol Sci 2023; 24:ijms24076730. [PMID: 37047701 PMCID: PMC10095444 DOI: 10.3390/ijms24076730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/27/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
Cartilage is an avascular tissue and sensitive to mechanical trauma and/or age-related degenerative processes leading to the development of osteoarthritis (OA). Therefore, it is important to investigate the mesenchymal cell-based chondrogenic regenerating mechanisms and possible their regulation. The aim of this study was to investigate the role of intracellular calcium (iCa2+) and its regulation through voltage-operated calcium channels (VOCC) on chondrogenic differentiation of mesenchymal stem/stromal cells derived from human bone marrow (BMMSCs) and menstrual blood (MenSCs) in comparison to OA chondrocytes. The level of iCa2+ was highest in chondrocytes, whereas iCa2+ store capacity was biggest in MenSCs and they proliferated better as compared to other cells. The level of CaV1.2 channels was also highest in OA chondrocytes than in other cells. CaV1.2 antagonist nifedipine slightly suppressed iCa2+, Cav1.2 and the proliferation of all cells and affected iCa2+ stores, particularly in BMMSCs. The expression of the CaV1.2 gene during 21 days of chondrogenic differentiation was highest in MenSCs, showing the weakest chondrogenic differentiation, which was stimulated by the nifedipine. The best chondrogenic differentiation potential showed BMMSCs (SOX9 and COL2A1 expression); however, purposeful iCa2+ and VOCC regulation by blockers can stimulate a chondrogenic response at least in MenSCs.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| |
Collapse
|
11
|
A novel implant surface modification mode of Fe3O4-containing TiO2 nanorods with sinusoidal electromagnetic field for osteoblastogenesis and angiogenesis. Mater Today Bio 2023; 19:100590. [PMID: 36910272 PMCID: PMC9996442 DOI: 10.1016/j.mtbio.2023.100590] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Implants made of Ti and its alloys are widely utilized in orthopaedic surgeries. However, insufficient osseointegration of the implants often causes complications such as aseptic loosening. Our previous research discovered that disordered titanium dioxide nanorods (TNrs) had satisfactory antibacterial properties and biocompatibility, but TNrs harmed angiogenic differentiation, which might retarded the osseointegration process of the implants. Magnetic nanomaterials have a certain potential in promoting osseointegration, electromagnetic fields within a specific frequency and intensity range can facilitate angiogenic and osteogenic differentiation. Therefore, this study used Fe3O4 to endow magnetism to TNrs and explored the regulation effects of Ti, TNrs, and Fe3O4-TNrs under 1 mT 15 Hz sinusoidal electromagnetic field (SEMF) on osteoblastogenesis, osseointegration, angiogenesis, and its mechanism. We discovered that after the addition of SEMF treatment to VR-EPCs cultured on Fe3O4-TNrs, the calcineurin/NFAT signaling pathway was activated, which then reversed the inhibitory effect of Fe3O4-TNrs on angiogenesis. Besides, Fe3O4-TNrs with SEMF enhanced osteogenic differentiation and osseointegration. Therefore, the implant modification mode of Fe3O4-TNrs with the addition of SEMF could more comprehensively promote osseointegration and provided a new idea for the modification of implants.
Collapse
|
12
|
Lee IC, Lin YC, Liu HL, Liu NC. Dual-frequency ultrasound enhances functional neuron differentiation from neural stem cells by modulating Ca 2+ dynamics and the ERK1/2 signaling pathway. J Cell Physiol 2023; 238:137-150. [PMID: 36350183 DOI: 10.1002/jcp.30911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
Our previous study demonstrated that ultrasound is able to promote differentiation on neural stem cells (NSCs), and dual-frequency ultrasound promotes this effect due to enhanced acoustic cavitation compared with single-frequency ultrasound. However, the underlying biological reasons have not been well disclosed. The purpose of this study was to investigate the underlying bioeffects, mechanisms and signaling pathways of dual-frequency ultrasound on NSC differentiation. The morphology, neurite outgrowth, and differentiation percentages were investigated under various dual-frequency simulation parameters with exposure periods varying from 5 to 15 min. Morphological observations identified that dual-frequency ultrasound stimulation promoted ultrasound dose-dependent neurite outgrowth. In particular, cells exposed for 10 min/2 days showed optimal neurite outgrowth and neuron differentiation percentages. In addition, live cell calcium images showed that dual-frequency ultrasound enhanced the internal calcium content of the cells, and calcium ions entering cells from the extracellular environment could be observed. Dual frequency ultrasound exposure enhanced extracellular calcium influx and upregulated extracellular signal-regulated kinases 1/2 (ERK1/2) expression. Observations from immunostaining and protein expression examinations also identified that dual-frequency ultrasound promoted brain-derived neurotrophic factor (BDNF) secretion from astrocytes derived from NSCs. In summary, evidence supports that dual-frequency ultrasound effectively enhances functional neuron differentiation via calcium channel regulation via the downstream ERK1/2 pathway and promotes BDNF secretion to serve as feedback to cascade neuron differentiation. The results may provide an alternative for cell-based therapy in brain injury.
Collapse
Affiliation(s)
- I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chieh Lin
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Nien-Che Liu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
13
|
Maione AS, Faris P, Iengo L, Catto V, Bisonni L, Lodola F, Negri S, Casella M, Guarino A, Polvani G, Cerrone M, Tondo C, Pompilio G, Sommariva E, Moccia F. Ca 2+ dysregulation in cardiac stromal cells sustains fibro-adipose remodeling in Arrhythmogenic Cardiomyopathy and can be modulated by flecainide. J Transl Med 2022; 20:522. [PMID: 36371290 PMCID: PMC9652790 DOI: 10.1186/s12967-022-03742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/30/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Cardiac mesenchymal stromal cells (C-MSC) were recently shown to differentiate into adipocytes and myofibroblasts to promote the aberrant remodeling of cardiac tissue that characterizes arrhythmogenic cardiomyopathy (ACM). A calcium (Ca2+) signaling dysfunction, mainly demonstrated in mouse models, is recognized as a mechanism impacting arrhythmic risk in ACM cardiomyocytes. Whether similar mechanisms influence ACM C-MSC fate is still unknown. Thus, we aim to ascertain whether intracellular Ca2+ oscillations and the Ca2+ toolkit are altered in human C-MSC obtained from ACM patients, and to assess their link with C-MSC-specific ACM phenotypes. METHODS AND RESULTS ACM C-MSC show enhanced spontaneous Ca2+ oscillations and concomitant increased Ca2+/Calmodulin dependent kinase II (CaMKII) activation compared to control cells. This is manly linked to a constitutive activation of Store-Operated Ca2+ Entry (SOCE), which leads to enhanced Ca2+ release from the endoplasmic reticulum through inositol-1,4,5-trisphosphate receptors. By targeting the Ca2+ handling machinery or CaMKII activity, we demonstrated a causative link between Ca2+ oscillations and fibro-adipogenic differentiation of ACM C-MSC. Genetic silencing of the desmosomal gene PKP2 mimics the remodelling of the Ca2+ signalling machinery occurring in ACM C-MSC. The anti-arrhythmic drug flecainide inhibits intracellular Ca2+ oscillations and fibro-adipogenic differentiation by selectively targeting SOCE. CONCLUSIONS Altogether, our results extend the knowledge of Ca2+ dysregulation in ACM to the stromal compartment, as an etiologic mechanism of C-MSC-related ACM phenotypes. A new mode of action of flecainide on a novel mechanistic target is unveiled against the fibro-adipose accumulation in ACM.
Collapse
Affiliation(s)
- Angela S Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy.
| | - Pawan Faris
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Lara Iengo
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
| | - Valentina Catto
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Luca Bisonni
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
| | - Francesco Lodola
- Laboratory of Cardiac Cellular Physiology, Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Sharon Negri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Michela Casella
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Cardiology and Arrhythmology Clinic, University Hospital "Umberto I-Salesi-Lancisi", Ancona, Italy
| | - Anna Guarino
- Cardiovascular Tissue Bank of Lombardy, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gianluca Polvani
- Cardiovascular Tissue Bank of Lombardy, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Marina Cerrone
- Medicine, Leon H. Charney Division of Cardiology, Heart Rhythm Center and Cardiovascular Genetics Program, New York University School of Medicine, New York, USA
| | - Claudio Tondo
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dentist Sciences, University of Milano, Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dentist Sciences, University of Milano, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138, Milan, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
14
|
Abdel Aziz I, Maver L, Giannasi C, Niada S, Brini AT, Antognazza MR. Polythiophene-mediated light modulation of membrane potential and calcium signalling in human adipose-derived stem/stromal cells. JOURNAL OF MATERIALS CHEMISTRY. C 2022; 10:9823-9833. [PMID: 36277082 PMCID: PMC9487879 DOI: 10.1039/d2tc01426b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/05/2022] [Indexed: 06/16/2023]
Abstract
Recent progress in the fields of regenerative medicine and tissue engineering has been strongly fostered both by the investigation of crucial cues, able to trigger the regeneration of damaged tissues, and by the development of ad hoc functional materials, capable of selectively (re-)activating relevant physiological pathways. In parallel to the successful realization of biochemical cues and the optimization of delivery protocols, the use of biophysical stimuli has been emerging as an alternative, highly effective strategy. Techniques based on electrical, magnetic and mechanical stimulation have been reported to efficiently direct differentiation of stem cells and modulate cell physiology at different developmental stages. In this framework, the use of optical stimulation represents a valuable approach, possibly overcoming current limitations of chemical cues, like limited spatial and temporal resolution and poor control over the extracellular environment. Surprisingly, the effects of light on the physiological properties (light toxicity, cell membrane potential, and cell ionic trafficking) of undifferentiated cells, as well as on their differentiation pathways, were investigated to a very limited extent and rarely quantified in a systematic way. In this work, we aim at clarifying the effects of optical excitation on the physiological behaviour of undifferentiated human adipose-derived stem cells (hASC), cultured on top of a light-sensitive conjugated polymer, region-regular poly-3-hexyl-thiophene (P3HT). Interestingly, we observe statistically significant modulation of the cell membrane potential, as well as noticeable effects on intracellular calcium signalling, triggered by P3HT excitation upon green light stimuli. Possible mechanisms involved in the signal transduction pathways are considered and critically discussed. The capability to modulate the physiological response of hASC upon photoexcitation, in a highly controlled and selective manner, provides a promptly available and non invasive diagnostic tool, thus contributing to the understanding of the complex machinery behind stem cells and material interfaces. Moreover, it may open the route to novel techniques to drive the differentiation path with unprecedented versatility and operational easiness.
Collapse
Affiliation(s)
- Ilaria Abdel Aziz
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3 20133 Milano Italy
- Politecnico di Milano, Dip.to di Fisica, P.zza L. da Vinci 32 20133 Milano Italy
| | - Leonardo Maver
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3 20133 Milano Italy
- Politecnico di Milano, Dip.to di Fisica, P.zza L. da Vinci 32 20133 Milano Italy
| | - Chiara Giannasi
- University of Milan, Department of Biomedical, Surgical and Dental Sciences, Via Vanvitelli 32 20129 Milano Italy
- IRCCS Istituto Ortopedico Galeazzi, Via Galeazzi 4 20161 Milano Italy
| | - Stefania Niada
- IRCCS Istituto Ortopedico Galeazzi, Via Galeazzi 4 20161 Milano Italy
| | - Anna T Brini
- University of Milan, Department of Biomedical, Surgical and Dental Sciences, Via Vanvitelli 32 20129 Milano Italy
- IRCCS Istituto Ortopedico Galeazzi, Via Galeazzi 4 20161 Milano Italy
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3 20133 Milano Italy
| |
Collapse
|
15
|
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
|
16
|
Faris P, Casali C, Negri S, Iengo L, Biggiogera M, Maione AS, Moccia F. Nicotinic Acid Adenine Dinucleotide Phosphate Induces Intracellular Ca2+ Signalling and Stimulates Proliferation in Human Cardiac Mesenchymal Stromal Cells. Front Cell Dev Biol 2022; 10:874043. [PMID: 35392169 PMCID: PMC8980055 DOI: 10.3389/fcell.2022.874043] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/24/2022] [Indexed: 12/18/2022] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a newly discovered second messenger that gates two pore channels 1 (TPC1) and 2 (TPC2) to elicit endo-lysosomal (EL) Ca2+ release. NAADP-induced lysosomal Ca2+ release may be amplified by the endoplasmic reticulum (ER) through the Ca2+-induced Ca2+ release (CICR) mechanism. NAADP-induced intracellular Ca2+ signals were shown to modulate a growing number of functions in the cardiovascular system, but their occurrence and role in cardiac mesenchymal stromal cells (C-MSCs) is still unknown. Herein, we found that exogenous delivery of NAADP-AM induced a robust Ca2+ signal that was abolished by disrupting the lysosomal Ca2+ store with Gly-Phe β-naphthylamide, nigericin, and bafilomycin A1, and blocking TPC1 and TPC2, that are both expressed at protein level in C-MSCs. Furthermore, NAADP-induced EL Ca2+ release resulted in the Ca2+-dependent recruitment of ER-embedded InsP3Rs and SOCE activation. Transmission electron microscopy revealed clearly visible membrane contact sites between lysosome and ER membranes, which are predicted to provide the sub-cellular framework for lysosomal Ca2+ to recruit ER-embedded InsP3Rs through CICR. NAADP-induced EL Ca2+ mobilization via EL TPC was found to trigger the intracellular Ca2+ signals whereby Fetal Bovine Serum (FBS) induces C-MSC proliferation. Furthermore, NAADP-evoked Ca2+ release was required to mediate FBS-induced extracellular signal-regulated kinase (ERK), but not Akt, phosphorylation in C-MSCs. These finding support the notion that NAADP-induced TPC activation could be targeted to boost proliferation in C-MSCs and pave the way for future studies assessing whether aberrant NAADP signaling in C-MSCs could be involved in cardiac disorders.
Collapse
Affiliation(s)
- Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Claudio Casali
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Lara Iengo
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Angela Serena Maione
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- *Correspondence: Angela Serena Maione, ; Francesco Moccia,
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
- *Correspondence: Angela Serena Maione, ; Francesco Moccia,
| |
Collapse
|
17
|
George LF, Bates EA. Mechanisms Underlying Influence of Bioelectricity in Development. Front Cell Dev Biol 2022; 10:772230. [PMID: 35237593 PMCID: PMC8883286 DOI: 10.3389/fcell.2022.772230] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/07/2022] [Indexed: 12/25/2022] Open
Abstract
To execute the intricate process of development, cells coordinate across tissues and organs to determine where each cell divides and differentiates. This coordination requires complex communication between cells. Growing evidence suggests that bioelectrical signals controlled via ion channels contribute to cell communication during development. Ion channels collectively regulate the transmembrane potential of cells, and their function plays a conserved role in the development of organisms from flies to humans. Spontaneous calcium oscillations can be found in nearly every cell type and tissue, and disruption of these oscillations leads to defects in development. However, the mechanism by which bioelectricity regulates development is still unclear. Ion channels play essential roles in the processes of cell death, proliferation, migration, and in each of the major canonical developmental signaling pathways. Previous reviews focus on evidence for one potential mechanism by which bioelectricity affects morphogenesis, but there is evidence that supports multiple different mechanisms which are not mutually exclusive. Evidence supports bioelectricity contributing to development through multiple different mechanisms. Here, we review evidence for the importance of bioelectricity in morphogenesis and provide a comprehensive review of the evidence for several potential mechanisms by which ion channels may act in developmental processes.
Collapse
Affiliation(s)
- Laura Faith George
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Emily Anne Bates
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
18
|
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]
|
19
|
McDonough RC, Price C. Targeted Activation of GPCR-Mediated Ca 2+ Signaling Drives Enhanced Cartilage-Like Matrix Formation. Tissue Eng Part A 2021; 28:405-419. [PMID: 34693731 PMCID: PMC9271335 DOI: 10.1089/ten.tea.2021.0078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Intracellular calcium ([Ca2+]i) signaling is a critical regulator of chondrogenesis, chondrocyte differentiation, and cartilage development. Calcium (Ca2+) signaling is known to direct processes that govern chondrocyte gene expression, protein synthesis, cytoskeletal remodeling, and cell fate. Control of chondrocyte/chondroprogenitor Ca2+ signaling has been attempted through mechanical and/or pharmacological activation of endogenous Ca2+ signaling transducers; however, such approaches can lack specificity and/or precision regarding Ca2+ activation mechanisms. Synthetic signaling platforms permitting precise and selective Ca2+ signal transduction can improve dissection of the roles that [Ca2+]i signaling play in chondrocyte behavior. One such platform is the chemogenetic hM3Dq DREADD (designer receptor exclusively activated by designer drugs) that activates [Ca2+]i signaling via the Gαq-PLCβ-IP3-ER pathway upon clozapine N-oxide (CNO) administration. We previously demonstrated hM3Dq's ability to precisely and synthetically initiate robust [Ca2+]i transients and oscillatory [Ca2+]i signaling in chondrocyte-like ATDC5 cells. Here, we investigate the effects that long-term CNO stimulatory culture have on hM3Dq [Ca2+]i signaling dynamics, proliferation, and protein deposition in 2D ATDC5 cultures. Long-term culturing under repeated CNO stimulation modified the temporal dynamics of hM3Dq [Ca2+]i signaling, increased cell proliferation, and enhanced matrix production in a CNO dose- and frequency-dependent manner, and triggered the formation of cell condensations that developed aligned, anisotropic neotissue structures rich in cartilaginous proteoglycans and collagens, all in the absence of differentiation inducers. This study demonstrated Gαq-GPCR-mediated [Ca2+]i signaling involvement in chondroprogenitor proliferation and cartilage-like matrix production, and established hM3Dq as a powerful tool for elucidating the role of GPCR-mediated Ca2+ signaling in chondrogenesis and chondrocyte differentiation.
Collapse
Affiliation(s)
- Ryan C McDonough
- University of Delaware, 5972, Biomedical Engineering, 161 Colburn Lab, Newark, Delaware, United States, 19716-5600;
| | - Christopher Price
- University of Delaware, 5972, Biomedical Engineering, Newark, Delaware, United States;
| |
Collapse
|
20
|
Torre EC, Bicer M, Cottrell GS, Widera D, Tamagnini F. Time-Dependent Reduction of Calcium Oscillations in Adipose-Derived Stem Cells Differentiating towards Adipogenic and Osteogenic Lineage. Biomolecules 2021; 11:biom11101400. [PMID: 34680033 PMCID: PMC8533133 DOI: 10.3390/biom11101400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived mesenchymal stromal cells (ASCs) are multipotent stem cells which can differentiate into various cell types, including osteocytes and adipocytes. Due to their ease of harvesting, multipotency, and low tumorigenicity, they are a prime candidate for the development of novel interventional approaches in regenerative medicine. ASCs exhibit slow, spontaneous Ca2+ oscillations and the manipulation of Ca2+ signalling via electrical stimulation was proposed as a potential route for promoting their differentiation in vivo. However, the effects of differentiation-inducing treatments on spontaneous Ca2+ oscillations in ASCs are not yet fully characterised. In this study, we used 2-photon live Ca2+ imaging to assess the fraction of cells showing spontaneous oscillations and the frequency of the oscillation (measured as interpeak interval—IPI) in ASCs undergoing osteogenic or adipogenic differentiation, using undifferentiated ASCs as controls. The measurements were carried out at 7, 14, and 21 days in vitro (DIV) to assess the effect of time in culture on Ca2+ dynamics. We observed that both time and differentiation treatment are important factors associated with a reduced fraction of cells showing Ca2+ oscillations, paralleled by increased IPI times, in comparison with untreated ASCs. Both adipogenic and osteogenic differentiation resulted in a reduction in Ca2+ dynamics, such as the fraction of cells showing intracellular Ca2+ oscillations and their frequency. Adipogenic differentiation was associated with a more pronounced reduction of Ca2+ dynamics compared to cells differentiating towards the osteogenic fate. Changes in Ca2+ associated oscillations with a specific treatment had already occurred at 7 DIV. Finally, we observed a reduction in Ca2+ dynamics over time in untreated ASCs. These data suggest that adipogenic and osteogenic differentiation cell fates are associated with specific changes in spontaneous Ca2+ dynamics over time. While this observation is interesting and provides useful information to understand the functional correlates of stem cell differentiation, further studies are required to clarify the molecular and mechanistic correlates of these changes. This will allow us to better understand the causal relationship between Ca2+ dynamics and differentiation, potentially leading to the development of novel, more effective interventions for both bone regeneration and control of adipose growth.
Collapse
Affiliation(s)
- Enrico C. Torre
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; (E.C.T.); (M.B.)
- Neuronal and Cellular Physiology Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
- Biomedicine West Wing, International Centre for Life, Times Square, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Mesude Bicer
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; (E.C.T.); (M.B.)
- Department of Bioengineering, Sumer Campus, Abdullah Gül University, Kayseri 38080, Turkey
| | - Graeme S. Cottrell
- Cellular and Molecular Neuroscience, School of Pharmacy, University of Reading, Reading RG6 6LA, UK;
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; (E.C.T.); (M.B.)
- Correspondence: (D.W.); (F.T.)
| | - Francesco Tamagnini
- Neuronal and Cellular Physiology Group, School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
- Correspondence: (D.W.); (F.T.)
| |
Collapse
|
21
|
Kongthitilerd P, Sharma A, Guidry HE, Rong W, Nguyen J, Yao S, Adisakwattana S, Cheng H. Antidiuretic hormone inhibits osteogenic differentiation of dental follicle stem cells via V1a receptors and the PLC-IP 3 pathway. Arch Oral Biol 2021; 128:105169. [PMID: 34058720 DOI: 10.1016/j.archoralbio.2021.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The aim of this study was to elucidate the molecular mechanism by which antidiuretic hormone (ADH) inhibited osteogenesis in dental follicle stem cells. DESIGN Rat dental follicle stem cells were cultured in osteogenic differentiation medium supplemented with ADH. Alkaline phosphatase enzyme activity, Alizarin Red S staining, MTT assay and RT-qPCR was used to examine ADH's impact on cell mineralization, viability, and osteogenic gene expression. Real-time calcium imaging analysis was performed to identify the ADH receptor and its mechanism of action. RESULTS ADH supplementation to the osteogenic differentiation medium inhibited cell mineralization without compromising cell viability and downregulated the expression of key osteogenic genes: DCN (Decorin), RUNX2 (Runt-related transcription factor 2) and BSP (Bone sialoprotein). Real-time calcium imaging analysis revealed that ADH (1-1000 nM) increased intracellular calcium in a concentration-dependent manner. Pretreatment of cells with V2255, a V1a receptor blocker, inhibited the calcium signals, but not with the V1b (Nelivaptan) or V2 (Tolvaptan). V2255 also reversed the inhibitory effect of ADH on osteogenesis. Furthermore, U73122, a Phospholipase C (PLC) inhibitor, 2-APB, an Inositol Triphosphate (IP3) receptor blocker, and depletion of endoplasmic reticulum calcium stores abolished the calcium signals by ADH. CONCLUSIONS Our results demonstrated that ADH activates V1a receptors and the PLC-IP3 pathway to stimulate intracellular calcium signals, which inhibits cell mineralization and osteogenic gene expression. These findings uncovered a novel function for ADH as a negative regulator of osteogenesis in dental follicle stem cells. The role of ADH in the pathogenesis of bone diseases remains to be determined.
Collapse
Affiliation(s)
- P Kongthitilerd
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA; Interdisciplinary Program of Biomedical Sciences, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - A Sharma
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - H E Guidry
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - W Rong
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - J Nguyen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - S Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - S Adisakwattana
- Phytochemical and Functional Food Research Unit for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - H Cheng
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
22
|
Ahamad N, Singh BB. Calcium channels and their role in regenerative medicine. World J Stem Cells 2021; 13:260-280. [PMID: 33959218 PMCID: PMC8080543 DOI: 10.4252/wjsc.v13.i4.260] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/22/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cells hold indefinite self-renewable capability that can be differentiated into all desired cell types. Based on their plasticity potential, they are divided into totipotent (morula stage cells), pluripotent (embryonic stem cells), multipotent (hematopoietic stem cells, multipotent adult progenitor stem cells, and mesenchymal stem cells [MSCs]), and unipotent (progenitor cells that differentiate into a single lineage) cells. Though bone marrow is the primary source of multipotent stem cells in adults, other tissues such as adipose tissues, placenta, amniotic fluid, umbilical cord blood, periodontal ligament, and dental pulp also harbor stem cells that can be used for regenerative therapy. In addition, induced pluripotent stem cells also exhibit fundamental properties of self-renewal and differentiation into specialized cells, and thus could be another source for regenerative medicine. Several diseases including neurodegenerative diseases, cardiovascular diseases, autoimmune diseases, virus infection (also coronavirus disease 2019) have limited success with conventional medicine, and stem cell transplantation is assumed to be the best therapy to treat these disorders. Importantly, MSCs, are by far the best for regenerative medicine due to their limited immune modulation and adequate tissue repair. Moreover, MSCs have the potential to migrate towards the damaged area, which is regulated by various factors and signaling processes. Recent studies have shown that extracellular calcium (Ca2+) promotes the proliferation of MSCs, and thus can assist in transplantation therapy. Ca2+ signaling is a highly adaptable intracellular signal that contains several components such as cell-surface receptors, Ca2+ channels/pumps/exchangers, Ca2+ buffers, and Ca2+ sensors, which together are essential for the appropriate functioning of stem cells and thus modulate their proliferative and regenerative capacity, which will be discussed in this review.
Collapse
Affiliation(s)
- Nassem Ahamad
- School of Dentistry, UT Health Science Center San Antonio, San Antonio, TX 78257, United States
| | - Brij B Singh
- School of Dentistry, UT Health Science Center San Antonio, San Antonio, TX 78257, United States
| |
Collapse
|
23
|
McDonough RC, Gilbert RM, Gleghorn JP, Price C. Targeted Gq-GPCR activation drives ER-dependent calcium oscillations in chondrocytes. Cell Calcium 2021; 94:102363. [PMID: 33550208 DOI: 10.1016/j.ceca.2021.102363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 11/28/2022]
Abstract
The temporal dynamics of calcium signaling are critical regulators of chondrocyte homeostasis and chondrogenesis. Calcium oscillations regulate differentiation and anabolic processes in chondrocytes and their precursors. Attempts to control chondrocyte calcium signaling have been achieved through mechanical perturbations and synthetic ion channel modulators. However, such stimuli can lack both local and global specificity and precision when evoking calcium signals. Synthetic signaling platforms can more precisely and selectively activate calcium signaling, enabling improved dissection of the roles of intracellular calcium ([Ca2+]i) in chondrocyte behavior. One such platform is hM3Dq, a chemogenetic DREADD (Designer Receptors Exclusively Activated by Designer Drugs) that activates calcium signaling via the Gαq-PLCβ-IP3-ER pathway upon administration of clozapine N-oxide (CNO). We previously described the first-use of hM3Dq to precisely mediate targeted, synthetic calcium signals in chondrocyte-like ATDC5 cells. Here, we generated stably expressing hM3Dq-ATDC5 cells to investigate the dynamics of Gαq-GPCR calcium signaling in depth. CNO drove robust calcium responses in a temperature- and concentration-dependent (1 pM-100 μM) manner and elicited elevated levels of oscillatory calcium signaling above 10 nM. hM3Dq-mediated calcium oscillations in ATDC5 cells were reliant on ER calcium stores for both initiation and sustenance, and the downregulation and recovery dynamics of hM3Dq after CNO stimulation align with traditionally reported GPCR recycling kinetics. This study successfully generated a stable hM3Dq cell line to precisely drive Gαq-GPCR-mediated and ER-dependent oscillatory calcium signaling in ATDC5 cells and established a novel tool to elucidate the role that GPCR-mediated calcium signaling plays in chondrocyte biology, cartilage pathology, and cartilage tissue engineering.
Collapse
Affiliation(s)
- Ryan C McDonough
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Rachel M Gilbert
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, United States.
| |
Collapse
|
24
|
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
|
25
|
Mengqi S, Wen S, Boxin Z, Minni L, Yan Z, Qun W, Yumei Z. Micro/nano topography with altered nanotube diameter differentially trigger endoplasmic reticulum stress to mediate bone mesenchymal stem cell osteogenic differentiation. Biomed Mater 2020; 16:015024. [PMID: 33036006 DOI: 10.1088/1748-605x/abbfee] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Micro/nano-topography (MNT) can promote osteogenic differentiation of stem cells, but the mechanism of topographical signaling transduction remains unclear. We have confirmed MNT, as a stressor, triggers endoplasmic reticulum (ER) stress and activates unfolded protein response in rat bone marrow mesenchymal stem cells, and such topography-induced ER stress promotes osteogenic differentiation. In order to reveal the influence of nanotube dimensions on ER stress, MNTs containing vertically oriented TiO2 nanotubes of diameters ranging from 30 nm to 100 nm were fabricated on pure titanium (Ti) foils, and ER stress and osteogenic differentiation of cells were systematically studied. After 12 h of cultivation, the transmission electron microscopy showed that cells on MNTs presented gross distortions of rough ER morphology containing the electron-dense material, and the expansion of the ER lumen became more pronounced as the dimension of nanotubes increased. Additionally, PCR and western blotting showed that the ER stress-related gene, the ER chaperone 78 kDa glucose-regulated protein, also known as binding-immunoglobulin protein (GRP78/BiP), was up-regulated, which was consistent with the osteogenesis-inducing ability of MNTs. Based on our previous studies, the findings in this article further revealed the mechanism for topographical cues modulating osteogenic differentiation of cells, which may provide an innovative approach for the optimal design of implant surface topography.
Collapse
Affiliation(s)
- Shi Mengqi
- Department of Stomatology, Navy Specialty Medical Center of Peoples' Liberation Army Navy, Shanghai 200052, People's Republic of China
- These authors contributed equally to this work
| | - Song Wen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, People's Republic of China
- These authors contributed equally to this work
| | - Zhang Boxin
- Department of Stomatology, Changzheng Hospital, the Second Military Medical University, Shanghai 200003, People's Republic of China
- These authors contributed equally to this work
| | - Liu Minni
- Department of Stomatology, Navy Specialty Medical Center of Peoples' Liberation Army Navy, Shanghai 200052, People's Republic of China
| | - Zhang Yan
- Department of Stomatology, Navy Specialty Medical Center of Peoples' Liberation Army Navy, Shanghai 200052, People's Republic of China
| | - Wu Qun
- Department of Stomatology, Navy Specialty Medical Center of Peoples' Liberation Army Navy, Shanghai 200052, People's Republic of China
| | - Zhang Yumei
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
26
|
Abstract
The purpose of this review is to highlight recent developments in the synthesis of chiral 1,4-dihydropyridines and their fused analogues. 1,4-Dihydropyridines are among the most active calcium antagonists that are used for the treatment of hypertension. Enantiomers of unsymmetrical 1,4-dihydropyridines often show different biological activities and may have even an opposite action profile. Hantzsch synthesis usually produces racemic mixtures of unsymmetrical 1,4-dihydropyridines. Therefore, the development of stereoselective synthesis of 1,4-dihydropyridines is one of the priorities of medicinal chemistry. Over the years, numerous methodologies have been developed for the production of enantiopure 1,4-dihydropyridines, such as stereoselective synthesis using chiral auxiliaries and chiral cyclocondensation partners, chromatographical methods, resolution of diastereomeric 1,4-dihydropyridine salts, enzyme catalysed kinetic resolution, or asymmetrisation of ester groups of 1,4-dihydropyridines. These approaches have been studied in detail and are relatively well established. The catalytic asymmetric approach holds the greatest promise in delivering the most practical and widely applicable methods. Substantial progress has been made toward the development of enantioselective organocatalytic methods for the construction of the chiral dihydropyridines. However, most of them do not provide a convenient way to pharmacologically important 1,4-dihydropyridine-3,5-dicarboxylates. Organocatalytic enantioselective desymmetrisation of prochiral 1,4-dihydropyridine-3,5-dicarbaldehydes also has great promise in the synthesis of pharmacologically important 1,4-dihydropyridine-3,5-dicarboxylates.
Collapse
|
27
|
Zhang L, Duan X, Sun W, Sun H. Perfluorooctane sulfonate acute exposure stimulates insulin secretion via GPR40 pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 726:138498. [PMID: 32305757 DOI: 10.1016/j.scitotenv.2020.138498] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 05/20/2023]
Abstract
Perfluoroalkyl substances (PFASs) are widely used synthetic chemicals, showing environmental/biological persistence and adverse effects on ecosystem and human health. Several epidemiological and animal studies have revealed that PFASs levels are associated with elevated serum insulin level; however, the effect of PFASs on insulin secretion and the underlying mechanism are not clear. In this study, the effect of a most concerned PFAS, perfluorooctane sulfonate (PFOS) on insulin secretion in Beta-TC-6 pancreatic cells was studied. The results showed that PFOS acute exposure stimulated insulin secretion and elevated intracellular calcium concentration ([Ca2+]i). The PFOS-stimulated [Ca2+]i elevation was resulted from both extra- and intra-cellular sources. PFOS acute exposure decreased ATP content and ATP/ADP ratio, indicating the mitochondrial function was damaged under PFOS acute exposure. The PFOS-stimulated insulin secretion was inhibited by GW1100, a G Protein-coupled Receptor 40 (GPR40) specific inhibitor, but not affected by GW9662, a specific antagonist to the peroxisome proliferator-activated receptor gamma (PPARγ). The observation of RNA silencing further demonstrated that the PFOS-stimulated insulin secretion is, at least partially, via GPR40. By using specific inhibitors, we found that the GPR40 downstream pathways, phospholipase C (PLC) and L-type Ca2+ channels (LTCC) were involved in PFOS-stimulated [Ca2+]i elevation and insulin secretion.
Collapse
Affiliation(s)
- Lianying Zhang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China; School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Xiaoyu Duan
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Weijie Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Hongwen Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| |
Collapse
|
28
|
Chen C, Hao X, Geng Z, Wang Z. ITRAQ-based quantitative proteomic analysis of MG63 in response to HIF-1α inducers. J Proteomics 2020; 211:103558. [DOI: 10.1016/j.jprot.2019.103558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/07/2019] [Accepted: 10/18/2019] [Indexed: 01/18/2023]
|
29
|
Ross CL, Zhou Y, McCall CE, Soker S, Criswell TL. The Use of Pulsed Electromagnetic Field to Modulate Inflammation and Improve Tissue Regeneration: A Review. Bioelectricity 2019; 1:247-259. [PMID: 34471827 PMCID: PMC8370292 DOI: 10.1089/bioe.2019.0026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pulsed electromagnetic field (PEMF) is emerging as innovative treatment for regulation of inflammation, which could have significant effects on tissue regeneration. PEMF modulates inflammatory processes through the regulation of pro- and anti-inflammatory cytokine secretion during different stages of inflammatory response. Consistent outcomes in studies involving animal and human tissue have shown promise for the use of PEMF as an alternative or complementary treatment to pharmaceutical therapies. Thus, PEMF treatment could provide a novel nonpharmaceutical means of modulating inflammation in injured tissues resulting in enhanced functional recovery. This review examines the effect of PEMF on immunomodulatory cells (e.g., mesenchymal stem/stromal cells [MSCs] and macrophages [MΦ]) to better understand the potential for PEMF therapy to modulate inflammatory signaling pathways and improve tissue regeneration. This review cites published data that support the use of PEMF to improve tissue regeneration. Our studies included herein confirm anti-inflammatory effects of PEMF on MSCs and MΦ.
Collapse
Affiliation(s)
- Christina L. Ross
- Center for Integrative Medicine, Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Yu Zhou
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Charles E. McCall
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Shay Soker
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tracy L. Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
30
|
Ju Y, Ren X, Zhao S. Distal C-terminus of Ca v 1.2 is indispensable for the chondrogenic differentiation of rat dental pulp stem cells. Cell Biol Int 2019; 44:512-523. [PMID: 31631478 DOI: 10.1002/cbin.11251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
The α1 subunit (Cav1.2) of the L-type calcium channel (LTCC), which is presently existing in both excitatory cells and non-excitatory cells, is involved in the differentiation and proliferation of mesenchymal stem cells (MSCs). Dental pulp stem cells (DPSCs), MSCs derived from dental pulp, exhibit multipotent characteristics similar to those of MSCs. The aim of the present study was to examine the contribution of Cav1.2 and its distal C-terminus (DCT) to the commitment of rat DPSCs (rDPSCs) toward chondrocytes and adipocytes in vitro. The expression of Cav1.2 was obviously elevated in chondrogenic differentiation but did not differ significantly in adipogenic differentiation. The chondrogenic differentiation but not adipogenic of rDPSCs was inhibited by either blocking LTCC using nimodipine or knockdown of Cav1.2 via short hairpin RNA (shRNA). Overexpression of DCT rescued the inhibition by Cav1.2-shRNA during chondrogenic differentiation, indicating that DCT is essential for the chondrogenic differentiation of rDPSCs. However, the protein level of DCT decreased after chondrogenic differentiation in wild-type cells, and overexpression of DCT in rDPSCs inhibited the phenotype. These data suggest that DCT is indispensable for chondrogenic differentiation of rDPSCs but that superfluous DCT inhibits this process. Through the analysis of differentially expressed genes using RNA-seq data, we speculated that the regulation of DCT might be mediated by the mitogen-activated protein kinase/extracellular-regulated kinase and c-Jun N-terminal kinase signaling pathways, or Chondromodulin-1.
Collapse
Affiliation(s)
- Yanqin Ju
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, P.R. China
| | - Xudong Ren
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, P.R. China
| | - Shouliang Zhao
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, 200040, P.R. China
| |
Collapse
|
31
|
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
|
32
|
Stovall KE, Tran TDN, Suantawee T, Yao S, Gimble JM, Adisakwattana S, Cheng H. Adenosine triphosphate enhances osteoblast differentiation of rat dental pulp stem cells via the PLC-IP 3 pathway and intracellular Ca 2+ signaling. J Cell Physiol 2019; 235:1723-1732. [PMID: 31301074 DOI: 10.1002/jcp.29091] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/11/2019] [Indexed: 01/01/2023]
Abstract
Intracellular Ca2+ signals are essential for stem cell function and play a significant role in the differentiation process. Dental pulp stem cells (DPSCs) are a potential source of stem cells; however, the mechanisms controlling cell differentiation remain largely unknown. Utilizing rat DPSCs, we examined the effect of adenosine triphosphate (ATP) on osteoblast differentiation and characterized its mechanism of action using real-time Ca 2+ imaging analysis. Our results revealed that ATP enhanced osteogenesis as indicated by Ca 2+ deposition in the extracellular matrix via Alizarin Red S staining. This was consistent with upregulation of osteoblast genes BMP2, Mmp13, Col3a1, Ctsk, Flt1, and Bgn. Stimulation of DPSCs with ATP (1-300 µM) increased intracellular Ca 2+ signals in a concentration-dependent manner, whereas histamine, acetylcholine, arginine vasopressin, carbachol, and stromal-cell-derived factor-1α failed to do so. Depletion of intracellular Ca 2+ stores in the endoplasmic reticulum by thapsigargin abolished the ATP responses which, nevertheless, remained detectable under extracellular Ca 2+ free condition. Furthermore, the phospholipase C (PLC) inhibitor U73122 and the inositol triphosphate (IP 3 ) receptor inhibitor 2-aminoethoxydiphenyl borate inhibited the Ca 2+ signals. Our findings provide a better understanding of how ATP controls osteogenesis in DPSCs, which involves a Ca 2+ -dependent mechanism via the PLC-IP 3 pathway. This knowledge could help improve osteogenic differentiation protocols for tissue regeneration of bone structures.
Collapse
Affiliation(s)
- Kelsie E Stovall
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Tran D N Tran
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Tanyawan Suantawee
- Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Jeffrey M Gimble
- LaCell LLC, New Orleans Bioinnovation Center, New Orleans, Louisiana.,Center for Stem Cell Research & Regenerative Medicine, Tulane University, New Orleans, Louisiana
| | - Sirichai Adisakwattana
- Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Henrique Cheng
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
33
|
Zhou S, Chen S, Jiang Q, Pei M. Determinants of stem cell lineage differentiation toward chondrogenesis versus adipogenesis. Cell Mol Life Sci 2019; 76:1653-1680. [PMID: 30689010 PMCID: PMC6456412 DOI: 10.1007/s00018-019-03017-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/10/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Adult stem cells, also termed as somatic stem cells, are undifferentiated cells, detected among differentiated cells in a tissue or an organ. Adult stem cells can differentiate toward lineage specific cell types of the tissue or organ in which they reside. They also have the ability to differentiate into mature cells of mesenchymal tissues, such as cartilage, fat and bone. Despite the fact that the balance has been comprehensively scrutinized between adipogenesis and osteogenesis and between chondrogenesis and osteogenesis, few reviews discuss the relationship between chondrogenesis and adipogenesis. In this review, the developmental and transcriptional crosstalk of chondrogenic and adipogenic lineages are briefly explored, followed by elucidation of signaling pathways and external factors guiding lineage determination between chondrogenic and adipogenic differentiation. An in-depth understanding of overlap and discrepancy between these two mesenchymal tissues in lineage differentiation would benefit regeneration of high-quality cartilage tissues and adipose tissues for clinical applications.
Collapse
Affiliation(s)
- Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, 610083, Sichuan, People's Republic of China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- Robert C. Byrd Health Sciences Center, WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
34
|
TRPV4-mediated calcium signaling in mesenchymal stem cells regulates aligned collagen matrix formation and vinculin tension. Proc Natl Acad Sci U S A 2019; 116:1992-1997. [PMID: 30674675 DOI: 10.1073/pnas.1811095116] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Microarchitectural cues drive aligned fibrillar collagen deposition in vivo and in biomaterial scaffolds, but the cell-signaling events that underlie this process are not well understood. Utilizing a multicellular patterning model system that allows for observation of intracellular signaling events during collagen matrix assembly, we investigated the role of calcium (Ca2+) signaling in human mesenchymal stem cells (MSCs) during this process. We observed spontaneous Ca2+ oscillations in MSCs during fibrillar collagen assembly, and hypothesized that the transient receptor potential vanilloid 4 (TRPV4) ion channel, a mechanosensitive Ca2+-permeable channel, may regulate this signaling. Inhibition of TRPV4 nearly abolished Ca2+ signaling at initial stages of collagen matrix assembly, while at later times had reduced but significant effects. Importantly, blocking TRPV4 activity dramatically reduced aligned collagen fibril assembly; conversely, activating TRPV4 accelerated aligned collagen formation. TRPV4-dependent Ca2+ oscillations were found to be independent of pattern shape or subpattern cell location, suggesting this signaling mechanism is necessary for aligned collagen formation but not sufficient in the absence of physical (microarchitectural) cues that force multicellular alignment. As cell-generated mechanical forces are known to be critical to the matrix assembly process, we examined the role of TRPV4-mediated Ca2+ signaling in force generated across the load-bearing focal adhesion protein vinculin within MSCs using an FRET-based tension sensor. Inhibiting TRPV4 decreased tensile force across vinculin, whereas TRPV4 activation caused a dynamic unloading and reloading of vinculin. Together, these findings suggest TRPV4 activity regulates forces at cell-matrix adhesions and is critical to aligned collagen matrix assembly by MSCs.
Collapse
|
35
|
Hou J, Luo T, Chen S, Lin S, Yang MM, Li G, Sun D. Calcium Spike Patterns Reveal Linkage of Electrical Stimulus and MSC Osteogenic Differentiation. IEEE Trans Nanobioscience 2019; 18:3-9. [PMID: 30442614 DOI: 10.1109/tnb.2018.2881004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) are easily obtained multipotent cells that are widely applied in regenerative medicine. Electrical stimulation (ES) has a promoting effect on bone healing and osteogenic differentiation of MSCs. Direct and alternating currents (AC) are extensively used to promote the osteogenic differentiation of MSCs in vivo and in vitro. However, information on conducting effective differentiation remains scarce. In this paper, we propose a method to optimize ES parameters based on calcium spike patterns of MSCs. Calcium spike frequency decreases as the osteogenic differentiation of MSC progresses. Furthermore, we tested various ES parameters through the real-time monitoring of calcium spike patterns. We efficiently initiated the process of osteogenic differentiation in MSCs by using the optimal parameters of AC, including voltage, signal shapes, frequency, and duty time. This method provides a new approach to optimize osteogenic differentiation and is potentially useful in clinical treatment such as of bone fractures.
Collapse
|
36
|
Uzieliene I, Bernotas P, Mobasheri A, Bernotiene E. The Role of Physical Stimuli on Calcium Channels in Chondrogenic Differentiation of Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19102998. [PMID: 30275359 PMCID: PMC6212952 DOI: 10.3390/ijms19102998] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/04/2018] [Accepted: 09/22/2018] [Indexed: 12/15/2022] Open
Abstract
Human mesenchymal stem cells (hMSC) are becoming increasingly popular in tissue engineering. They are the most frequently used stem cell source for clinical applications due to their high potential to differentiate into several lineages. Cartilage is known for its low capacity for self-maintenance and currently there are no efficient methods to improve cartilage repair. Chondrogenic differentiation of hMSC isolated from different tissues is widely employed due to a high clinical demand for the improvement of cartilage regeneration. Calcium channels that are regulated by physical stimuli seem to play a pivotal role in chondrogenic differentiation of MSCs. These channels increase intracellular calcium concentration, which leads to the initiation of the relevant cellular processes that are required for differentiation. This review will focus on the impact of different physical stimuli, including electrical, electromagnetic/magnetic and mechanical on various calcium channels and calcium signaling mechanisms during chondrogenic differentiation of hMSC.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania.
| | - Paulius Bernotas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania.
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania.
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania.
| |
Collapse
|
37
|
Razlivanov I, Liew T, Moore EW, Al-Kathiri A, Bartram T, Kuvshinov D, Nikolaev A. Long-term imaging of calcium dynamics using genetically encoded calcium indicators and automatic tracking of cultured cells. Biotechniques 2018; 65:37-39. [PMID: 30014737 DOI: 10.2144/btn-2018-0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Calcium dynamics is crucial for many signaling pathways and cell functions. Understanding how calcium regulates cell function often requires long-term imaging of calcium dynamics. Here we report a methodological approach of long-term (5-10 h) imaging of calcium dynamics in cultured cells. The approach links calcium imaging using genetically encoded calcium indicators and semi-automatic tracking of individual cells. It can be used in a large variety of situations, ranging from the role of calcium in biological processes to cell heterogeneity and screening of drugs modifying signaling pathways.
Collapse
Affiliation(s)
- Igor Razlivanov
- Department of Biomedical Sciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Teresa Liew
- Department of Biomedical Sciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Eira Watts Moore
- Department of Biomedical Sciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Alaa Al-Kathiri
- Department of Biomedical Sciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Tayma Bartram
- Department of Biomedical Sciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.,Department of Oncology and Metabolism, The University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Dmitriy Kuvshinov
- Department of Chemical Engineering, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Anton Nikolaev
- Department of Biomedical Sciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
38
|
Ermakov A, Daks A, Fedorova O, Shuvalov O, Barlev NA. Ca 2+ -depended signaling pathways regulate self-renewal and pluripotency of stem cells. Cell Biol Int 2018; 42:1086-1096. [PMID: 29851182 DOI: 10.1002/cbin.10998] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/25/2018] [Indexed: 12/15/2022]
Abstract
Ca2+ -mediated signaling is widely spread in nature and plays critical role in the individual development of various organisms ranging from microorganisms to mammals. In vertebrates, Ca2+ is involved in important developmental events: fertilization, body plan establishment, and organogenesis. The two later events are defined by embryonic stem cells (ESCs). ESCs are capable of self-renewal and are pluripotent by nature, that is, can give rise to all types of cells that make up the body. Given the paramount importance of Ca2+ signalization in the development, it is therefore not surprising this process also plays role in the biology of stem cells. In this review, we scrutinize the published experimental data on the role of Ca2+ ions in embryonic stem cells self-renewal and pluripotency. In line with this, we also discuss possible mechanisms of p53 inhibition as a major hindrance to self-renewal of ESCs. Finally, we argue about the role of G-protein-coupled receptors (GPCRs), the largest family of heteromeric transmembrane receptors, and GPCR-mediated signalization in stem cells, and propose the role for the GPCR-G-protein-PLC-Ca2+ -downstream signaling pathway in the regulation of pluripotency of both mouse and human ESCs.
Collapse
Affiliation(s)
| | - Alexandra Daks
- Institute of Cytology RAS, Saint-Petersburg 194064, Russia
| | - Olga Fedorova
- Institute of Cytology RAS, Saint-Petersburg 194064, Russia
| | - Oleg Shuvalov
- Institute of Cytology RAS, Saint-Petersburg 194064, Russia
| | | |
Collapse
|
39
|
Malenczyk K, Szodorai E, Schnell R, Lubec G, Szabó G, Hökfelt T, Harkany T. Secretagogin protects Pdx1 from proteasomal degradation to control a transcriptional program required for β cell specification. Mol Metab 2018; 14:108-120. [PMID: 29910119 PMCID: PMC6034064 DOI: 10.1016/j.molmet.2018.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Specification of endocrine cell lineages in the developing pancreas relies on extrinsic signals from non-pancreatic tissues, which initiate a cell-autonomous sequence of transcription factor activation and repression switches. The steps in this pathway share reliance on activity-dependent Ca2+ signals. However, the mechanisms by which phasic Ca2+ surges become converted into a dynamic, cell-state-specific and physiologically meaningful code made up by transcription factors constellations remain essentially unknown. METHODS We used high-resolution histochemistry to explore the coincident expression of secretagogin and transcription factors driving β cell differentiation. Secretagogin promoter activity was tested in response to genetically manipulating Pax6 and Pax4 expression. Secretagogin null mice were produced with their pancreatic islets morphologically and functionally characterized during fetal development. A proteomic approach was utilized to identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome and verified in vitro by focusing on Pdx1 retention. RESULTS Here, we show that secretagogin, a Ca2+ sensor protein that controls α and β cell turnover in adult, is in fact expressed in endocrine pancreas from the inception of lineage segregation in a Pax4-and Pax6-dependent fashion. By genetically and pharmacologically manipulating secretagogin expression and interactome engagement in vitro, we find secretagogin to gate excitation-driven Ca2+ signals for β cell differentiation and insulin production. Accordingly, secretagogin-/- fetuses retain a non-committed pool of endocrine progenitors that co-express both insulin and glucagon. We identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome complex to prevent Pdx1 degradation through proteasome inactivation. This coincides with retained Nkx6.1, Pax4 and insulin transcription in prospective β cells. CONCLUSIONS In sum, secretagogin scales the temporal availability of a Ca2+-dependent transcription factor network to define β cell identity.
Collapse
Affiliation(s)
- Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Paracelsus Medical University, Strubergasse 21, A-5020, Salzburg, Austria
| | - Robert Schnell
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, SE-17177, Stockholm, Sweden
| | - Gert Lubec
- Paracelsus Medical University, Strubergasse 21, A-5020, Salzburg, Austria
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony utca 43, H-1083, Budapest, Hungary
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden.
| |
Collapse
|
40
|
Kunrath-Lima M, de Miranda MC, Ferreira ADF, Faraco CCF, de Melo MIA, Goes AM, Rodrigues MA, Faria JAQA, Gomes DA. Phospholipase C delta 4 (PLCδ4) is a nuclear protein involved in cell proliferation and senescence in mesenchymal stromal stem cells. Cell Signal 2018; 49:59-67. [PMID: 29859928 DOI: 10.1016/j.cellsig.2018.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 02/08/2023]
Abstract
Ca2+ is an important second messenger, and it is involved in many cellular processes such as cell death and proliferation. The rise in intracellular Ca2+ levels can be due to the generation of inositol 1,4,5-trisphosphate (InsP3), which is a product of phosphatidylinositol 4,5-bisphosphate (PIP2) hydrolysis by phospholipases C (PLCs), that leads to Ca2+ release from endoplasmic reticulum by InsP3 receptors (InsP3R). Ca2+ signaling patterns can vary in different regions of the cell and increases in nuclear Ca2+ levels have specific biological effects that differ from those of Ca2+ increase in the cytoplasm. There are PLCs in the cytoplasm and nucleus, but little is known about the functions of nuclear PLCs. This work aimed to characterize phenotypically the human PLCδ4 (hPLCδ4) in mesenchymal stem cells. This nuclear isoform of PLC is present in different cell types and has a possible role in proliferative processes. In this work, hPLCδ4 was found to be mainly nuclear in human adipose-derived mesenchymal stem cells (hASC). PLCδ4 knockdown demonstrated that it is essential for hASC proliferation, without inducing cell death. An increase of cells in G1, and a reduction of cells on interphase and G2/M in knockdown cells were seen. Furthermore, PLCδ4 knockdown increased the percentage of senescent cells, p16INK4A+ and p21Cip1 mRNAs expression, which could explain the impaired cell proliferation. The results show that hPLCδ4 is in involved in cellular proliferation and senescence in hASC.
Collapse
Affiliation(s)
- Marianna Kunrath-Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcelo Coutinho de Miranda
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Andrea da Fonseca Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Camila Cristina Fraga Faraco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mariane Izabella Abreu de Melo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alfredo Miranda Goes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Michele Angela Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Dawidson Assis Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
41
|
Lee SH, Rigas NK, Lee CR, Bang A, Srikanth S, Gwack Y, Kang MK, Kim RH, Park NH, Shin KH. Orai1 promotes tumor progression by enhancing cancer stemness via NFAT signaling in oral/oropharyngeal squamous cell carcinoma. Oncotarget 2017; 7:43239-43255. [PMID: 27259269 PMCID: PMC5190020 DOI: 10.18632/oncotarget.9755] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 01/03/2023] Open
Abstract
Emerging evidence indicates that Orai1, a key calcium channel for store-operated Ca2+ entry, is associated with human cancer. However, the underlying mechanism by which Orai1 regulates cancer progression remains unknown. Here we report that intracellular level of Orai1 is increased in a stepwise manner during oral/oropharyngeal carcinogenesis and highly expressed in cancer stem-like cell (CSC)-enriched populations of human oral/oropharyngeal squamous cell carcinoma (OSCC). Ectopic Orai1 expression converted non-tumorigenic immortalized oral epithelial cells to malignant cells that showed CSC properties, e.g., self-renewal capacity, increased ALDH1HIGH cell population, increased key stemness transcription factors, and enhanced mobility. Conversely, inhibition of Orai1 suppressed tumorigenicity and CSC phenotype of OSCC, indicating that Orai1 could be an important element for tumorigenicity and stemness of OSCC. Mechanistically, Orai1 activates its major downstream effector molecule, NFATc3. Knockdown of NFATc3 in the Orai1-overexpressing oral epithelial cells abrogates the effect of Orai1 on CSC phenotype. Moreover, antagonist of NFAT signaling also decreases CSC phenotype, implying the functional importance of Orai1/NFAT axis in OSCC CSC regulation. Our study identifies Orai1 as a novel molecular determinant for OSCC progression by enhancing cancer stemness, suggesting that inhibition of Orai1 signaling may offer an effective therapeutic modality against OSCC.
Collapse
Affiliation(s)
- Sung Hee Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Nicole Kristina Rigas
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Chang-Ryul Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - April Bang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mo K Kang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
42
|
Altered spontaneous calcium signaling of in situ chondrocytes in human osteoarthritic cartilage. Sci Rep 2017; 7:17093. [PMID: 29213100 PMCID: PMC5719003 DOI: 10.1038/s41598-017-17172-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/20/2017] [Indexed: 12/23/2022] Open
Abstract
Intracellular calcium ([Ca2+]i) signaling is an essential universal secondary messenger in articular chondrocytes. However, little is known about its spatiotemporal features in the context of osteoarthritis (OA). Herein, by examining the cartilage samples collected from patients undergoing knee arthroscopic surgery, we investigated the spatiotemporal features of spontaneous [Ca2+]i signaling in in situ chondrocytes at different OA stages. Our data showed zonal dependent spontaneous [Ca2+]i signaling in healthy cartilage samples under 4 mM calcium environment. This signal was significantly attenuated in healthy cartilage samples but increased in early-degenerated cartilage when cultured in 0 mM calcium environment. No significant difference was found in [Ca2+]i intensity oscillation in chondrocytes located in middle zones among ICRS 1–3 samples under both 4 and 0 mM calcium environments. However, the correlation was found in deep zone chondrocytes incubated in 4 mM calcium environment. In addition, increased protein abundance of Cav3.3 T-type voltage dependent calcium channel and Nfatc2 activity were observed in early-degenerated cartilage samples. The present study exhibited OA severity dependent spatiotemporal features of spontaneous [Ca2+]i oscillations of in situ chondrocytes, which might reflect the zonal specific role of chondrocytes during OA progression and provide new insight in articular cartilage degradation during OA progression.
Collapse
|
43
|
Comparative Analysis of Spontaneous and Stimulus-Evoked Calcium Transients in Proliferating and Differentiating Human Midbrain-Derived Stem Cells. Stem Cells Int 2017; 2017:9605432. [PMID: 29201062 PMCID: PMC5671755 DOI: 10.1155/2017/9605432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/10/2017] [Indexed: 12/24/2022] Open
Abstract
Spontaneous cytosolic calcium transients and oscillations have been reported in various tissues of nonhuman and human origin but not in human midbrain-derived stem cells. Using confocal microfluorimetry, we studied spontaneous calcium transients and calcium-regulating mechanisms in a human ventral mesencephalic stem cell line undergoing proliferation and neuronal differentiation. Spontaneous calcium transients were detected in a large fraction of both proliferating (>50%) and differentiating (>55%) cells. We provide evidence for the existence of intracellular calcium stores that respond to muscarinic activation of the cells, having sensitivity for ryanodine and thapsigargin possibly reflecting IP3 receptor activity and the presence of ryanodine receptors and calcium ATPase pumps. The observed calcium transient activity potentially supports the existence of a sodium-calcium antiporter and the existence of calcium influx induced by depletion of calcium stores. We conclude that the cells have developed the most important mechanisms governing cytosolic calcium homeostasis. This is the first comparative report of spontaneous calcium transients in proliferating and differentiating human midbrain-derived stem cells that provides evidence for the mechanisms that are likely to be involved. We propose that the observed spontaneous calcium transients may contribute to mechanisms involved in cell proliferation, phenotypic differentiation, and general cell maturation.
Collapse
|
44
|
Kang P, Kumar S, Schaffer D. Novel biomaterials to study neural stem cell mechanobiology and improve cell-replacement therapies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 4:13-20. [PMID: 29399646 PMCID: PMC5791915 DOI: 10.1016/j.cobme.2017.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neural stem cells (NSCs) are a valuable cell source for tissue engineering, regenerative medicine, disease modeling, and drug screening applications. Analogous to other stem cells, NSCs are tightly regulated by their microenvironmental niche, and prior work utilizing NSCs as a model system with engineered biomaterials has offered valuable insights into how biophysical inputs can regulate stem cell proliferation, differentiation, and maturation. In this review, we highlight recent exciting studies with innovative material platforms that enable narrow stiffness gradients, mechanical stretching, temporal stiffness switching, and three-dimensional culture to study NSCs. These studies have significantly advanced our knowledge of how stem cells respond to an array of different biophysical inputs and the underlying mechanosensitive mechanisms. In addition, we discuss efforts to utilize engineered material scaffolds to improve NSC-based translational efforts and the importance of mechanobiology in tissue engineering applications.
Collapse
Affiliation(s)
- Phillip Kang
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Lawrence Berkeley National Laboratory Physical Biosciences Division, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - David Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
45
|
McLeod C, Mauck R. On the origin and impact of mesenchymal stem cell heterogeneity: new insights and emerging tools for single cell analysis. Eur Cell Mater 2017; 34:217-231. [PMID: 29076514 PMCID: PMC7735381 DOI: 10.22203/ecm.v034a14] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) display substantial cell-to-cell variation. This heterogeneity manifests among donors, among tissue sources, and within cell populations. Such pervasive variability complicates the use of MSCs in regenerative applications and may limit their therapeutic efficacy. Most conventional assays measure MSC properties in bulk and, as a consequence, mask this cell-to-cell variation. Recent studies have identified extensive variability amongst and within clonal MSC populations, in dimensions including functional differentiation capacity, molecular state (e.g. epigenetic, transcriptomic, and proteomic status), and biophysical properties. While the origins of these variations remain to be elucidated, potential mechanisms include in vivo micro-anatomical heterogeneity, epigenetic bistability, and transcriptional fluctuations. Emerging tools for single cell analysis of MSC gene and protein expression may yield further insight into the mechanisms and implications of single cell variation amongst these cells, and ultimately improve the clinical utility of MSCs in tissue engineering and regenerative medicine applications. This review outlines the dimensions across which MSC heterogeneity is present, defines some of the known mechanisms that govern this heterogeneity, and highlights emerging technologies that may further refine our understanding and improve our clinical application of this unique cell type.
Collapse
Affiliation(s)
- C.M. McLeod
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA,McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, PA 19104, USA
| | - R.L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA,McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, PA 19104, USA.,Address for correspondence: Robert L. Mauck, PhD, McKay Orthopaedic Research Laboratory, University of Pennsylvania, 424 Stemmler Hall, 36th Street and Hamilton Walk, Philadelphia, PA 19104, USA, Telephone: 1-215-898-3294 FAX: 1-215-573-2133
| |
Collapse
|
46
|
Pchelintseva E, Djamgoz MBA. Mesenchymal stem cell differentiation: Control by calcium-activated potassium channels. J Cell Physiol 2017; 233:3755-3768. [PMID: 28776687 DOI: 10.1002/jcp.26120] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in modern medicine for which understanding the mechanisms controlling their differentiation is fundamental. Ion channels offer novel insights to this process because of their role in modulating membrane potential and intracellular milieu. Here, we evaluate the contribution of calcium-activated potassium (KCa ) channels to the three main components of MSC differentiation: initiation, proliferation, and migration. First, we demonstrate the importance of the membrane potential (Vm ) and the apparent association of hyperpolarization with differentiation. Of KCa subtypes, most evidence points to activity of big-conductance channels in inducing initiation. On the other hand, intermediate-conductance currents have been shown to promote progression through the cell cycle. While there is no information on the role of KCa channels in migration of MSCs, work from other stem cells and cancer cells suggest that intermediate-conductance and to a lesser extent big-conductance channels drive migration. In all cases, these effects depend on species, tissue origin and lineage. Finally, we present a conceptual model that demonstrates how KCa activity could influence differentiation by regulating Vm and intracellular Ca2+ oscillations. We conclude that KCa channels have significant involvement in MSC differentiation and could potentially enable novel tissue engineering approaches and therapies.
Collapse
Affiliation(s)
- Ekaterina Pchelintseva
- Department of Life Sciences, Imperial College London, South Kensington Campus, Neuroscience Solution to Cancer Research Group, London, UK.,Department of Bioengineering, Imperial College London, South Kensington Campus, London, UK
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, Neuroscience Solution to Cancer Research Group, London, UK
| |
Collapse
|
47
|
Parate D, Franco-Obregón A, Fröhlich J, Beyer C, Abbas AA, Kamarul T, Hui JHP, Yang Z. Enhancement of mesenchymal stem cell chondrogenesis with short-term low intensity pulsed electromagnetic fields. Sci Rep 2017; 7:9421. [PMID: 28842627 PMCID: PMC5572790 DOI: 10.1038/s41598-017-09892-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/28/2017] [Indexed: 12/22/2022] Open
Abstract
Pulse electromagnetic fields (PEMFs) have been shown to recruit calcium-signaling cascades common to chondrogenesis. Here we document the effects of specified PEMF parameters over mesenchymal stem cells (MSC) chondrogenic differentiation. MSCs undergoing chondrogenesis are preferentially responsive to an electromagnetic efficacy window defined by field amplitude, duration and frequency of exposure. Contrary to conventional practice of administering prolonged and repetitive exposures to PEMFs, optimal chondrogenic outcome is achieved in response to brief (10 minutes), low intensity (2 mT) exposure to 6 ms bursts of magnetic pulses, at 15 Hz, administered only once at the onset of chondrogenic induction. By contrast, repeated exposures diminished chondrogenic outcome and could be attributed to calcium entry after the initial induction. Transient receptor potential (TRP) channels appear to mediate these aspects of PEMF stimulation, serving as a conduit for extracellular calcium. Preventing calcium entry during the repeated PEMF exposure with the co-administration of EGTA or TRP channel antagonists precluded the inhibition of differentiation. This study highlights the intricacies of calcium homeostasis during early chondrogenesis and the constraints that are placed on PEMF-based therapeutic strategies aimed at promoting MSC chondrogenesis. The demonstrated efficacy of our optimized PEMF regimens has clear clinical implications for future regenerative strategies for cartilage.
Collapse
Affiliation(s)
- Dinesh Parate
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore, 119288, Singapore
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, IE Kent Ridge Road, Singapore, 119228, Singapore. .,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 medical Drive, #14-01, Singapore, 117599, Singapore.
| | - Jürg Fröhlich
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, IE Kent Ridge Road, Singapore, 119228, Singapore.,Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Rämistrasse 101, 8092, Zurich, Switzerland
| | - Christian Beyer
- Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Rämistrasse 101, 8092, Zurich, Switzerland
| | - Azlina A Abbas
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Pantai Valley, Kuala Lumpur, 50603, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Pantai Valley, Kuala Lumpur, 50603, Malaysia
| | - James H P Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore, 119288, Singapore. .,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510, Singapore.
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore, 119288, Singapore. .,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510, Singapore.
| |
Collapse
|
48
|
Cyanidin Stimulates Insulin Secretion and Pancreatic β-Cell Gene Expression through Activation of l-type Voltage-Dependent Ca 2+ Channels. Nutrients 2017; 9:nu9080814. [PMID: 28788070 PMCID: PMC5579608 DOI: 10.3390/nu9080814] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
Cyanidin is a natural anthocyanidin present in fruits and vegetables with anti-diabetic properties including stimulation of insulin secretion. However, its mechanism of action remains unknown. In this study, we elucidated the mechanisms of cyanidin for stimulatory insulin secretion from pancreatic β-cells. Rat pancreatic β-cells INS-1 were used to investigate the effects of cyanidin on insulin secretion, intracellular Ca2+ signaling, and gene expression. We detected the presence of cyanidin in the intracellular space of β-cells. Cyanidin stimulated insulin secretion and increased intracellular Ca2+ signals in a concentration-dependent manner. The Ca2+ signals were abolished by nimodipine, an l-type voltage-dependent Ca2+ channel (VDCC) blocker or under extracellular Ca2+ free conditions. Stimulation of cells with cyanidin activated currents typical for VDCCs and up-regulated the expression of glucose transporter 2 (GLUT2), Kir6.2, and Cav1.2 genes. Our findings indicate that cyanidin diffuses across the plasma membrane, leading to activation of l-type VDCCs. The increase in intracellular Ca2+ stimulated insulin secretion and the expression of genes involved in this process. These findings suggest that cyanidin could be used as a promising agent to stimulate insulin secretion.
Collapse
|
49
|
Nam HY, Balaji Raghavendran HR, Pingguan-Murphy B, Abbas AA, Merican AM, Kamarul T. Fate of tenogenic differentiation potential of human bone marrow stromal cells by uniaxial stretching affected by stretch-activated calcium channel agonist gadolinium. PLoS One 2017; 12:e0178117. [PMID: 28654695 PMCID: PMC5487029 DOI: 10.1371/journal.pone.0178117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/06/2017] [Indexed: 01/16/2023] Open
Abstract
The role for mechanical stimulation in the control of cell fate has been previously proposed, suggesting that there may be a role of mechanical conditioning in directing mesenchymal stromal cells (MSCs) towards specific lineage for tissue engineering applications. Although previous studies have reported that calcium signalling is involved in regulating many cellular processes in many cell types, its role in managing cellular responses to tensile loading (mechanotransduction) of MSCs has not been fully elucidated. In order to establish this, we disrupted calcium signalling by blocking stretch-activated calcium channel (SACC) in human MSCs (hMSCs) in vitro. Passaged-2 hMSCs were exposed to cyclic tensile loading (1 Hz + 8% for 6, 24, 48, and 72 hours) in the presence of the SACC blocker, gadolinium. Analyses include image observations of immunochemistry and immunofluorescence staining from extracellular matrix (ECM) production, and measuring related tenogenic and apoptosis gene marker expression. Uniaxial tensile loading increased the expression of tenogenic markers and ECM production. However, exposure to strain in the presence of 20 μM gadolinium reduced the induction of almost all tenogenic markers and ECM staining, suggesting that SACC acts as a mechanosensor in strain-induced hMSC tenogenic differentiation process. Although cell death was observed in prolonged stretching, it did not appear to be apoptosis mediated. In conclusion, the knowledge gained in this study by elucidating the role of calcium in MSC mechanotransduction processes, and that in prolonged stretching results in non-apoptosis mediated cell death may be potential useful for regenerative medicine applications.
Collapse
Affiliation(s)
- Hui Yin Nam
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail: (HYN); (TK)
| | - Hanumantha Rao Balaji Raghavendran
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Azlina A. Abbas
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Azhar M. Merican
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail: (HYN); (TK)
| |
Collapse
|
50
|
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
|