101
|
Niu Q, Shen S, He J, Wang L. CKIP-1 contributes to osteogenic differentiation of mouse bone marrow mesenchymal stem cells. Regen Med 2021; 16:847-859. [PMID: 34498492 DOI: 10.2217/rme-2020-0119] [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: 11/21/2022] Open
Abstract
Background: Osteogenesis greatly depends on the differentiation of bone marrow mesenchymal stem cells (BMSCs). CKIP-1 is considered to be a negative regulator of BMSCs. Methods: We established a CKIP-1 knockout mouse model, then isolated and cultured BMSCs from wild-type and knockout groups. Results: Our data demonstrated that CKIP-1 knockout significantly increased bone structure in the experimental mouse model and enhanced BMSC proliferation. CKIP-1 knockout contributed to osteoblastic and adipogenic differentiation. Furthermore, CKIP-1 regulated osteogenesis in BMSCs via the MAPK signaling pathway, and BMSCs from the CKIP-1 knockout mice were effective in repairing the skull defect null mice. Conclusion: Our results concluded that silencing of CKIP-1 promoted osteogenesis in experimental mice and increased BMSCs differentiation via upregulation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Qiannan Niu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Orthodontics, The Hospital of Stomatology, The Fourth Military Medical University, No.145 West Changle Road, Xi'an, 710000, Shaanxi, China
| | - Shuning Shen
- Department of Stomatology, No.984 Hospital of PLA, Beijing, 100094, China
| | - Jiaojiao He
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Orthodontics, The Hospital of Stomatology, The Fourth Military Medical University, No.145 West Changle Road, Xi'an, 710000, Shaanxi, China
| | - Lei Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Orthodontics, The Hospital of Stomatology, The Fourth Military Medical University, No.145 West Changle Road, Xi'an, 710000, Shaanxi, China
| |
Collapse
|
102
|
Morozumi W, Aoshima K, Inagaki S, Iwata Y, Nakamura S, Hara H, Shimazawa M. Piezo 1 is involved in intraocular pressure regulation. J Pharmacol Sci 2021; 147:211-221. [PMID: 34217619 DOI: 10.1016/j.jphs.2021.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/28/2021] [Accepted: 06/09/2021] [Indexed: 01/02/2023] Open
Abstract
Trabecular meshwork (TM) regulates the intraocular pressure (IOP) through the control of aqueous humor outflow. Previous reports show that TM cells express 11 types of mechanosensitive molecules, including Piezo 1, which sense mechanical stimuli. However, the role of Piezo 1 on TM remains unclear. Thus, in this study, we focused on the Piezo 1 and examined its role in TM cells. Immunostaining showed that Piezo 1 was expressed in mouse TM and human TM cells. Moreover, the eye drops containing Piezo 1 agonist Yoda 1 reduced the IOP in mice, and also reduced fibronectin expression level around the TM. In addition, Piezo 1 activation suppressed human TM cells migration/proliferation, and decreased fibronectin expression level. On the other hand, Piezo 1 activation increased matrix metalloproteinase (MMP)-2 expression responsible for fibronectin degradation. These findings could contribute to the development of new treatments for glaucoma.
Collapse
Affiliation(s)
- Wataru Morozumi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Kota Aoshima
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoshi Inagaki
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Iwata
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
103
|
Xu X, Liu S, Liu H, Ru K, Jia Y, Wu Z, Liang S, Khan Z, Chen Z, Qian A, Hu L. Piezo Channels: Awesome Mechanosensitive Structures in Cellular Mechanotransduction and Their Role in Bone. Int J Mol Sci 2021; 22:ijms22126429. [PMID: 34208464 PMCID: PMC8234635 DOI: 10.3390/ijms22126429] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 12/13/2022] Open
Abstract
Piezo channels are mechanosensitive ion channels located in the cell membrane and function as key cellular mechanotransducers for converting mechanical stimuli into electrochemical signals. Emerged as key molecular detectors of mechanical forces, Piezo channels' functions in bone have attracted more and more attention. Here, we summarize the current knowledge of Piezo channels and review the research advances of Piezo channels' function in bone by highlighting Piezo1's role in bone cells, including osteocyte, bone marrow mesenchymal stem cell (BM-MSC), osteoblast, osteoclast, and chondrocyte. Moreover, the role of Piezo channels in bone diseases is summarized.
Collapse
Affiliation(s)
- Xia Xu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Shuyu Liu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Hua Liu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Kang Ru
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yunxian Jia
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zixiang Wu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Shujing Liang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zarnaz Khan
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zhihao Chen
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Airong Qian
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Correspondence: (A.Q.); (L.H.)
| | - Lifang Hu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Correspondence: (A.Q.); (L.H.)
| |
Collapse
|
104
|
Rajakylä EK, Lehtimäki JI, Acheva A, Schaible N, Lappalainen P, Krishnan R, Tojkander S. Assembly of Peripheral Actomyosin Bundles in Epithelial Cells Is Dependent on the CaMKK2/AMPK Pathway. Cell Rep 2021; 30:4266-4280.e4. [PMID: 32209483 DOI: 10.1016/j.celrep.2020.02.096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 12/02/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
Defects in the maintenance of intercellular junctions are associated with loss of epithelial barrier function and consequent pathological conditions, including invasive cancers. Epithelial integrity is dependent on actomyosin bundles at adherens junctions, but the origin of these junctional bundles is incompletely understood. Here we show that peripheral actomyosin bundles can be generated from a specific actin stress fiber subtype, transverse arcs, through their lateral fusion at cell-cell contacts. Importantly, we find that assembly and maintenance of peripheral actomyosin bundles are dependent on the mechanosensitive CaMKK2/AMPK signaling pathway and that inhibition of this route leads to disruption of tension-maintaining actomyosin bundles and re-growth of stress fiber precursors. This results in redistribution of cellular forces, defects in monolayer integrity, and loss of epithelial identity. These data provide evidence that the mechanosensitive CaMKK2/AMPK pathway is critical for the maintenance of peripheral actomyosin bundles and thus dictates cell-cell junctions through cellular force distribution.
Collapse
Affiliation(s)
- Eeva Kaisa Rajakylä
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Niccole Schaible
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
105
|
Hendrickx G, Fischer V, Liedert A, von Kroge S, Haffner-Luntzer M, Brylka L, Pawlus E, Schweizer M, Yorgan T, Baranowsky A, Rolvien T, Neven M, Schumacher U, Beech DJ, Amling M, Ignatius A, Schinke T. Piezo1 Inactivation in Chondrocytes Impairs Trabecular Bone Formation. J Bone Miner Res 2021; 36:369-384. [PMID: 33180356 DOI: 10.1002/jbmr.4198] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/21/2020] [Accepted: 10/11/2020] [Indexed: 01/01/2023]
Abstract
The skeleton is a dynamic tissue continuously adapting to mechanical stimuli. Although matrix-embedded osteocytes are considered as the key mechanoresponsive bone cells, all other skeletal cell types are principally exposed to macroenvironmental and microenvironmental mechanical influences that could potentially affect their activities. It was recently reported that Piezo1, one of the two mechanically activated ion channels of the Piezo family, functions as a mechanosensor in osteoblasts and osteocytes. Here we show that Piezo1 additionally plays a critical role in the process of endochondral bone formation. More specifically, by targeted deletion of Piezo1 or Piezo2 in either osteoblast (Runx2Cre) or osteoclast lineage cells (Lyz2Cre), we observed severe osteoporosis with numerous spontaneous fractures specifically in Piezo1Runx2Cre mice. This phenotype developed at an early postnatal stage and primarily affected the formation of the secondary spongiosa. The presumptive Piezo1Runx2Cre osteoblasts in this region displayed an unusual flattened appearance and were positive for type X collagen. Moreover, transcriptome analyses of primary osteoblasts identified an unexpected induction of chondrocyte-related genes in Piezo1Runx2Cre cultures. Because Runx2 is not only expressed in osteoblast progenitor cells, but also in prehypertrophic chondrocytes, these data suggested that Piezo1 functions in growth plate chondrocytes to ensure trabecular bone formation in the process of endochondral ossification. To confirm this hypothesis, we generated mice with Piezo1 deletion in chondrocytes (Col2a1Cre). These mice essentially recapitulated the phenotype of Piezo1Runx2Cre animals, because they displayed early-onset osteoporosis with multiple fractures, as well as impaired formation of the secondary spongiosa with abnormal osteoblast morphology. Our data identify a previously unrecognized key function of Piezo1 in endochondral ossification, which, together with its role in bone remodeling, suggests that Piezo1 represents an attractive target for the treatment of skeletal disorders. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Gretl Hendrickx
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Laura Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Pawlus
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Department of Electron Microscopy, Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timur Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Baranowsky
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
106
|
Zhao YH, Wang JJ, Zhang PP, Hao HS, Pang YW, Wang HY, Du WH, Zhao SJ, Ruan WM, Zou HY, Hao T, Zhu HB, Zhao XM. Oocyte IVM or vitrification significantly impairs DNA methylation patterns in blastocysts as analysed by single-cell whole-genome methylation sequencing. Reprod Fertil Dev 2021; 32:676-689. [PMID: 32317092 DOI: 10.1071/rd19234] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
To explore the mechanisms leading to the poor quality of IVF blastocysts, the single-cell whole-genome methylation sequencing technique was used in this study to analyse the methylation patterns of bovine blastocysts derived from invivo, fresh (IVF) or vitrified (V_IVF) oocytes. Genome methylation levels of blastocysts in the IVF and V_IVF groups were significantly lower than those of the invivo group (P<0.05). In all, 1149 differentially methylated regions (DMRs) were identified between the IVF and invivo groups, 1578 DMRs were identified between the V_IVF and invivo groups and 151 DMRs were identified between the V_IVF and IVF groups. For imprinted genes, methylation levels of insulin-like growth factor 2 receptor (IGF2R) and protein phosphatase 1 regulatory subunit 9A (PPP1R9A) were lower in the IVF and V_IVF groups than in the invivo group, and the methylation level of paternally expressed 3 (PEG3) was lower in the V_IVF group than in the IVF and invivo groups. Genes with DMRs between the IVF and invivo and the V_IVF and IVF groups were primarily enriched in oocyte maturation pathways, whereas DMRs between the V_IVF and invivo groups were enriched in fertilisation and vitrification-vulnerable pathways. The results of this study indicate that differences in the methylation of critical DMRs may contribute to the differences in quality between invitro- and invivo-derived embryos.
Collapse
Affiliation(s)
- Ya-Han Zhao
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Jing-Jing Wang
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Pei-Pei Zhang
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Hai-Sheng Hao
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Yun-Wei Pang
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Hao-Yu Wang
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Wei-Hua Du
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Shan-Jiang Zhao
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Wei-Min Ruan
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Ming Lun Street, Kaifeng, Henan, 475004, PR China
| | - Hui-Ying Zou
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Tong Hao
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Hua-Bin Zhu
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China
| | - Xue-Ming Zhao
- Embryo Biotechnology and Reproduction Laboratory and the Centre of Domestic Animal Reproduction and Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, PR China; and Corresponding author.
| |
Collapse
|
107
|
A New Hope in Spinal Degenerative Diseases: Piezo1. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6645193. [PMID: 33575334 PMCID: PMC7857891 DOI: 10.1155/2021/6645193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/09/2021] [Indexed: 12/19/2022]
Abstract
As a newly discovered mechanosensitive ion channel protein, the piezo1 protein participates in the transmission of mechanical signals on the cell membrane and plays a vital role in mammalian biomechanics. Piezo1 has attracted widespread attention since it was discovered in 2010. In recent years, studies on piezo1 have gradually increased and deepened. In addition to the discovery that piezo1 is expressed in the respiratory, cardiovascular, gastrointestinal, and urinary systems, it is also stably expressed in cells such as mesenchymal stem cells (MSCs), osteoblasts, osteoclasts, chondrocytes, and nucleus pulposus cells that constitute vertebral bodies and intervertebral discs. They can all receive external mechanical stimulation through the piezo1 protein channel to affect cell proliferation, differentiation, migration, and apoptosis to promote the occurrence and development of lumbar degenerative diseases. Through reviewing the relevant literature of piezo1 in the abovementioned cells, this paper discusses the effect of piezo1 protein expression under mechanical stress stimuli on spinal degenerative disease, providing the molecular basis for the pathological mechanism of spinal degenerative disease and also a new basis, ideas, and methods for the prevention and treatment of this degenerative disease.
Collapse
|
108
|
He Y, Makarczyk MJ, Lin H. Role of mitochondria in mediating chondrocyte response to mechanical stimuli. Life Sci 2020; 263:118602. [PMID: 33086121 PMCID: PMC7736591 DOI: 10.1016/j.lfs.2020.118602] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/22/2020] [Accepted: 10/11/2020] [Indexed: 12/21/2022]
Abstract
As the most common form of arthritis, osteoarthritis (OA) has become a major cause of severe joint pain, physical disability, and quality of life impairment in the affected population. To date, precise pathogenesis of OA has not been fully clarified, which leads to significant obstacles in developing efficacious treatments such as failures in finding disease-modifying OA drugs (DMOADs) in the last decades. Given that diarthrodial joints primarily display the weight-bearing and movement-supporting function, it is not surprising that mechanical stress represents one of the major risk factors for OA. However, the inner connection between mechanical stress and OA onset/progression has yet to be explored. Mitochondrion, a widespread organelle involved in complex biological regulation processes such as adenosine triphosphate (ATP) synthesis and cellular metabolism, is believed to have a controlling role in the survival and function implement of chondrocytes, the singular cell type within cartilage. Mitochondrial dysfunction has also been observed in osteoarthritic chondrocytes. In this review, we systemically summarize mitochondrial alterations in chondrocytes during OA progression and discuss our recent progress in understanding the potential role of mitochondria in mediating mechanical stress-associated osteoarthritic alterations of chondrocytes. In particular, we propose the potential signaling pathways that may regulate this process, which provide new views and therapeutic targets for the prevention and treatment of mechanical stress-associated OA.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Meagan J Makarczyk
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
109
|
Pei F, Liu J, Zhang L, Pan X, Huang W, Cen X, Huang S, Jin Y, Zhao Z. The functions of mechanosensitive ion channels in tooth and bone tissues. Cell Signal 2020; 78:109877. [PMID: 33296740 DOI: 10.1016/j.cellsig.2020.109877] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 02/08/2023]
Abstract
Tooth and bone are independent tissues with a close relationship. Both are composed of a highly calcified outer structure and soft inner tissue, and both are constantly under mechanical stress. In particular, the alveolar bone and tooth constitute an occlusion system and suffer from masticatory and occlusal force. Thus, mechanotransduction is a key process in many developmental, physiological and pathological processes in tooth and bone. Mechanosensitive ion channels such as Piezo1 and Piezo2 are important participants in mechanotransduction, but their functions in tooth and bone are poorly understood. This review summarizes our current understanding of mechanosensitive ion channels and their roles in tooth and bone tissues. Research in these areas may shed new light on the regulation of tooth and bone tissues and potential treatments for diseases affecting these tissues.
Collapse
Affiliation(s)
- Fang Pei
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Jialing Liu
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Xuefeng Pan
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Wei Huang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China
| | - Xiao Cen
- Department of the Temporomandibular Joint, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Shishu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, PR China.
| | - Ying Jin
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, PR China.
| |
Collapse
|
110
|
Hasegawa K, Fujii S, Matsumoto S, Tajiri Y, Kikuchi A, Kiyoshima T. YAP signaling induces PIEZO1 to promote oral squamous cell carcinoma cell proliferation. J Pathol 2020; 253:80-93. [PMID: 32985688 DOI: 10.1002/path.5553] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/18/2020] [Accepted: 09/15/2020] [Indexed: 01/13/2023]
Abstract
Most cancer cells are exposed to altered extracellular environments, such as an increase in extracellular matrix (ECM) stiffness and soluble signals consisting of growth factors and cytokines. It is therefore conceivable that changes in tumor extracellular environments affect tumor cell behavior. The Hippo pathway reportedly responds to the extracellular environment and regulates the nuclear localization of the transcription co-activator, yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ). Inactivation of the Hippo pathway with nuclear translocation of YAP/TAZ stimulates cell proliferation. Its pathway also regulates gene expression, but the precise molecule(s) meditating the cell-proliferating effect of YAP signaling on oral squamous cell carcinoma (OSCC) is unclear. First, we examined the effects of YAP signaling on OSCC tumorigenesis. Loss-of-function experiments using siRNA or an inhibitor, and immunohistochemical analyses of tissue specimens obtained from OSCC patients demonstrated that YAP signaling was involved in OSCC cell proliferation. Second, we identified Piezo-type mechanosensitive ion channel component 1 (PIEZO1), a Ca2+ channel, as a transcriptional target of YAP signaling and showed that elevated PIEZO1 was required for PIEZO1 agonist-dependent Ca2+ entry and cell proliferation in OSCC cells. Experiments using three-dimensional and suspension culture revealed that PIEZO1 was involved in OSCC cellular growth. Finally, YAP overexpression in the nucleus and/or cytoplasm was immunohistochemically detected in tumor lesions with frequent expression of both PIEZO1 and Ki-67, but not in non-tumor regions of OSCC specimens. These results suggest that the YAP/PIEZO1 axis promotes OSCC cell growth. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kana Hasegawa
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shinsuke Fujii
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yudai Tajiri
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,Department of Dentistry and Oral Surgery, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
111
|
de Lucas B, Pérez LM, Bernal A, Gálvez BG. Ultrasound Therapy: Experiences and Perspectives for Regenerative Medicine. Genes (Basel) 2020; 11:genes11091086. [PMID: 32957737 PMCID: PMC7563547 DOI: 10.3390/genes11091086] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Ultrasound has emerged as a novel tool for clinical applications, particularly in the context of regenerative medicine. Due to its unique physico-mechanical properties, low-intensity ultrasound (LIUS) has been approved for accelerated fracture healing and for the treatment of established non-union, but its utility has extended beyond tissue engineering to other fields, including cell regeneration. Cells and tissues respond to acoustic ultrasound by switching on genetic repair circuits, triggering a cascade of molecular signals that promote cell proliferation, adhesion, migration, differentiation, and extracellular matrix production. LIUS also induces angiogenesis and tissue regeneration and has anti-inflammatory and anti-degenerative effects. Accordingly, the potential application of ultrasound for tissue repair/regeneration has been tested in several studies as a stand-alone treatment and, more recently, as an adjunct to cell-based therapies. For example, ultrasound has been proposed to improve stem cell homing to target tissues due to its ability to create a transitional and local gradient of cytokines and chemokines. In this review, we provide an overview of the many applications of ultrasound in clinical medicine, with a focus on its value as an adjunct to cell-based interventions. Finally, we discuss the various preclinical and clinical studies that have investigated the potential of ultrasound for regenerative medicine.
Collapse
Affiliation(s)
- Beatriz de Lucas
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
| | - Laura M. Pérez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
| | - Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - Beatriz G. Gálvez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
- Correspondence:
| |
Collapse
|
112
|
Orapiriyakul W, Tsimbouri MP, Childs P, Campsie P, Wells J, Fernandez-Yague MA, Burgess K, Tanner KE, Tassieri M, Meek D, Vassalli M, Biggs MJP, Salmeron-Sanchez M, Oreffo ROC, Reid S, Dalby MJ. Nanovibrational Stimulation of Mesenchymal Stem Cells Induces Therapeutic Reactive Oxygen Species and Inflammation for Three-Dimensional Bone Tissue Engineering. ACS NANO 2020; 14:10027-10044. [PMID: 32658450 PMCID: PMC7458485 DOI: 10.1021/acsnano.0c03130] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
There is a pressing clinical need to develop cell-based bone therapies due to a lack of viable, autologous bone grafts and a growing demand for bone grafts in musculoskeletal surgery. Such therapies can be tissue engineered and cellular, such as osteoblasts, combined with a material scaffold. Because mesenchymal stem cells (MSCs) are both available and fast growing compared to mature osteoblasts, therapies that utilize these progenitor cells are particularly promising. We have developed a nanovibrational bioreactor that can convert MSCs into bone-forming osteoblasts in two- and three-dimensional, but the mechanisms involved in this osteoinduction process remain unclear. Here, to elucidate this mechanism, we use increasing vibrational amplitude, from 30 nm (N30) to 90 nm (N90) amplitudes at 1000 Hz and assess MSC metabolite, gene, and protein changes. These approaches reveal that dose-dependent changes occur in MSCs' responses to increased vibrational amplitude, particularly in adhesion and mechanosensitive ion channel expression and that energetic metabolic pathways are activated, leading to low-level reactive oxygen species (ROS) production and to low-level inflammation as well as to ROS- and inflammation-balancing pathways. These events are analogous to those that occur in the natural bone-healing processes. We have also developed a tissue engineered MSC-laden scaffold designed using cells' mechanical memory, driven by the stronger N90 stimulation. These mechanistic insights and cell-scaffold design are underpinned by a process that is free of inductive chemicals.
Collapse
Affiliation(s)
- Wich Orapiriyakul
- Centre
for the Cellular Microenvironment, Institute of Molecular, Cell and
Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
- Department
of Orthopedics, Faculty of Medicine, Prince
of Songkla University, Songkhla 90110, Thailand
| | - Monica P. Tsimbouri
- Centre
for the Cellular Microenvironment, Institute of Molecular, Cell and
Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Peter Childs
- Centre
for the Cellular Microenvironment, Division of Biomedical Engineering,
School of Engineering, University of Glasgow, Glasgow G12 8LT, United Kingdom
| | - Paul Campsie
- SUPA
Department of Biomedical Engineering, University
of Strathclyde, Glasgow G1 1QE, United Kingdom
| | - Julia Wells
- Bone
and Joint Research Group, Centre for Human Development, Stem Cells
and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Marc A. Fernandez-Yague
- Centre for
Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Karl Burgess
- Glasgow
Polyomics, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Switchback Rd, Bearsden, Glasgow G61 1BD, United
Kingdom
| | - K. Elizabeth Tanner
- Centre
for the Cellular Microenvironment, Division of Biomedical Engineering,
School of Engineering, University of Glasgow, Glasgow G12 8LT, United Kingdom
- School
of Engineering and Materials Science, Queen
Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Manlio Tassieri
- Centre
for the Cellular Microenvironment, Division of Biomedical Engineering,
School of Engineering, University of Glasgow, Glasgow G12 8LT, United Kingdom
| | - Dominic Meek
- Department
of Orthopedics, Queen Elizabeth II University
Hospital, Glasgow G51 4TF, United Kingdom
| | - Massimo Vassalli
- Centre
for the Cellular Microenvironment, Division of Biomedical Engineering,
School of Engineering, University of Glasgow, Glasgow G12 8LT, United Kingdom
| | - Manus J. P. Biggs
- Centre for
Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Manuel Salmeron-Sanchez
- Centre
for the Cellular Microenvironment, Division of Biomedical Engineering,
School of Engineering, University of Glasgow, Glasgow G12 8LT, United Kingdom
| | - Richard O. C. Oreffo
- Bone
and Joint Research Group, Centre for Human Development, Stem Cells
and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Stuart Reid
- SUPA
Department of Biomedical Engineering, University
of Strathclyde, Glasgow G1 1QE, United Kingdom
| | - 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 G12 8QQ, United Kingdom
| |
Collapse
|
113
|
Mechanical regulation of cell size, fate, and behavior during asymmetric cell division. Curr Opin Cell Biol 2020; 67:9-16. [PMID: 32768924 DOI: 10.1016/j.ceb.2020.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/15/2020] [Accepted: 07/06/2020] [Indexed: 01/03/2023]
Abstract
Asymmetric cell division (ACD) is an evolutionary conserved mechanism used by prokaryotes and eukaryotes alike to generate cell diversity. ACD can be manifested in biased segregation of macromolecules or differential partitioning of cell organelles. Cells are also constantly subject to extrinsic or intrinsic mechanical forces, influencing cell behavior and fate. During ACD, cell intrinsic forces generated through the spatiotemporal regulation of the actomyosin cytoskeleton can influence sibling cell size. External mechanical stresses are further translated by transcriptional coactivators or mechanically gated ion channels. Here, we will discuss recent literature, exploring how mechanical cues influence various aspects of ACD and stem cell behavior, and how these mechanical cues contribute to cell fate decisions.
Collapse
|
114
|
Wu T, Yin F, Wang N, Ma X, Jiang C, Zhou L, Zong Y, Shan H, Xia W, Lin Y, Zhou Z, Yu X. Involvement of mechanosensitive ion channels in the effects of mechanical stretch induces osteogenic differentiation in mouse bone marrow mesenchymal stem cells. J Cell Physiol 2020; 236:284-293. [PMID: 32592173 DOI: 10.1002/jcp.29841] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/07/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) can be induced to process osteogenic differentiation with appropriate mechanical and/or chemical stimuli. The present study described the successful culture of murine BMSCs under mechanical strain. BMSCs were subjected to 0%, 3%, 8%, 13%, and 18% cyclic tensile strain at 0.5 Hz for 8 hr/day for 3 days. The expression of osteogenic markers and mechanosensitive ion channels was evaluated with real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blot. The expression of alkaline phosphatase (ALP) and matrix mineralization were evaluated with histochemical staining. To investigate the effects of mechanosensitive ion channel expression on cyclic tensile strain-induced osteogenic differentiation, the expression of osteogenic markers was evaluated with real-time RT-PCR in the cells without mechanosensitive ion channel expression. This study revealed a significant augment in osteogenic marker in BMSC strained at 8% compared to other treatments; therefore, an 8% strain was used for further investigations. The ALP expression and matrix mineralization were enhanced in osteogenic induced BMSCs subjected to 8% strain after 7 and 14 days, respectively. Under the same conditions, the osteogenic marker and mechanosensitive ion channel expression were significantly promoted. However, the loss function of mechanosensitive ion channels resulted in the inhibition of osteogenic marker expression. This study demonstrated that strain alone can successfully induce osteogenic differentiation in BMSCs and the expression of mechanosensitive ion channels was involved in the process. The current findings suggest that mechanical stretch could function as efficient stimuli to induce the osteogenic differentiation of BMSCs via the activation of mechanosensitive ion channels.
Collapse
Affiliation(s)
- Tianyi Wu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Fuli Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Nan Wang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Ma
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chaolai Jiang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lihui Zhou
- Department of Orthopaedic Surgery, Xiangshan First People's Hospital, Ningbo, Zhejiang, China
| | - Yang Zong
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenyang Xia
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zubin Zhou
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaowei Yu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
115
|
Felix L, Stephan J, Rose CR. Astrocytes of the early postnatal brain. Eur J Neurosci 2020; 54:5649-5672. [PMID: 32406559 DOI: 10.1111/ejn.14780] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
In the rodent forebrain, the majority of astrocytes are generated during the early postnatal phase. Following differentiation, astrocytes undergo maturation which accompanies the development of the neuronal network. Neonate astrocytes exhibit a distinct morphology and domain size which differs to their mature counterparts. Moreover, many of the plasma membrane proteins prototypical for fully developed astrocytes are only expressed at low levels at neonatal stages. These include connexins and Kir4.1, which define the low membrane resistance and highly negative membrane potential of mature astrocytes. Newborn astrocytes moreover express only low amounts of GLT-1, a glutamate transporter critical later in development. Furthermore, they show specific differences in the properties and spatio-temporal pattern of intracellular calcium signals, resulting from differences in their repertoire of receptors and signalling pathways. Therefore, roles fulfilled by mature astrocytes, including ion and transmitter homeostasis, are underdeveloped in the young brain. Similarly, astrocytic ion signalling in response to neuronal activity, a process central to neuron-glia interaction, differs between the neonate and mature brain. This review describes the unique functional properties of astrocytes in the first weeks after birth and compares them to later stages of development. We conclude that with an immature neuronal network and wider extracellular space, astrocytic support might not be as demanding and critical compared to the mature brain. The delayed differentiation and maturation of astrocytes in the first postnatal weeks might thus reflect a reduced need for active, energy-consuming regulation of the extracellular space and a less tight control of glial feedback onto synaptic transmission.
Collapse
Affiliation(s)
- Lisa Felix
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Jonathan Stephan
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
116
|
Bai WY, Wang L, Ying ZM, Hu B, Xu L, Zhang GQ, Cong PK, Zhu X, Zou W, Zheng HF. Identification of PIEZO1 polymorphisms for human bone mineral density. Bone 2020; 133:115247. [PMID: 31968281 DOI: 10.1016/j.bone.2020.115247] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/26/2022]
Abstract
Bone mineral density (BMD) is a key indicator for diagnosis and treatment for osteoporosis; the reduction of BMD could increase the risk of osteoporotic fracture. It was very recently found that Piezo1 mediated mechanically evoked responses in bone and further participated in bone formation in mice. Here, we performed cross phenotype meta-analysis for human BMD at lumbar spine (LS), femoral neck (FN), distal radius/forearm (FA) and heel and screened out 14 top SNPs for PIEZO1, these SNPs were overlapped with putative enhancers, DNase-I hypersensitive sites and active promoter flanking regions. We found that the signal of the best SNP rs62048221 was mainly from heel ultrasound estimated BMD (-0.02 SD per T allele, P = 8.50E-09), where calcaneus supported most of the mechanical force of body when standing, walking and doing physical exercises. Each copy of the effect allele T of SNP rs62048221 was associated with a decrease of 0.0035 g/cm2 BMD (P = 4.6E-27, SE = 0.0003) in UK Biobank data within 477,760 samples. SNP rs62048221 was located at the enhancer region (HEDD enhancer ID 2331049) of gene PIEZO1, site-directed ChIP assays in human mesenchymal stem cells (hMSCs) showed significant enrichment of H3K4me1 and H3K27ac in this region, luciferase assays showed that rs62048221 could significantly affect the activity of the enhancer where it resides. Our results first suggested that SNP rs62048221 might mediate the PIEZO1 expression level via modulating the activity of cis-regulatory elements and then further affect the BMD.
Collapse
Affiliation(s)
- Wei-Yang Bai
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, China
| | - Lijun Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhi-Min Ying
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Bin Hu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lin Xu
- Department of Orthopedics, The Affiliated Yantai Hospital of Binzhou Medical University, Yantai 264000, China
| | - Guo-Qing Zhang
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, China
| | - Pei-Kuan Cong
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, China
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Hou-Feng Zheng
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, China.
| |
Collapse
|
117
|
Song J, Liu L, Lv L, Hu S, Tariq A, Wang W, Dang X. Fluid shear stress induces Runx-2 expression via upregulation of PIEZO1 in MC3T3-E1 cells. Cell Biol Int 2020; 44:1491-1502. [PMID: 32181967 DOI: 10.1002/cbin.11344] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/14/2020] [Indexed: 01/14/2023]
Abstract
Mechanically induced biological responses in bone cells involve a complex biophysical process. Although various mechanosensors have been identified, the precise mechanotransduction pathway remains poorly understood. PIEZO1 is a newly discovered mechanically activated ion channel in bone cells. This study aimed to explore the involvement of PIEZO1 in mechanical loading (fluid shear stress)-induced signaling cascades that control osteogenesis. The results showed that fluid shear stress increased PIEZO1 expression in MC3T3-E1 cells. The fluid shear stress elicited the key osteoblastic gene Runx-2 expression; however, PIEZO1 silencing using small interference RNA blocked these effects. The AKT/GSK-3β/β-catenin pathway was activated in this process. PIEZO1 silencing impaired mechanically induced activation of the AKT/GSK-3β/β-catenin pathway. Therefore, the results demonstrated that MC3T3-E1 osteoblasts required PIEZO1 to adapt to the external mechanical fluid shear stress, thereby inducing osteoblastic Runx-2 gene expression, partly through the AKT/GSK-3β/β-catenin pathway.
Collapse
Affiliation(s)
- Jidong Song
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Liying Liu
- The Center Laboratory for Biomedical Research, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Leifeng Lv
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Shugang Hu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Alkhatatbeh Tariq
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Wei Wang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Xiaoqian Dang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| |
Collapse
|
118
|
The role of Piezo proteins and cellular mechanosensing in tuning the fate of transplanted stem cells. Cell Tissue Res 2020; 381:1-12. [DOI: 10.1007/s00441-020-03191-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
|
119
|
Shen Y, Pan Y, Guo S, Sun L, Zhang C, Wang L. The roles of mechanosensitive ion channels and associated downstream MAPK signaling pathways in PDLC mechanotransduction. Mol Med Rep 2020; 21:2113-2122. [PMID: 32323761 PMCID: PMC7115221 DOI: 10.3892/mmr.2020.11006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 02/05/2020] [Indexed: 12/30/2022] Open
Abstract
The present study aimed to investigate whether the cytoskeleton, the Piezo1 ion channel and the transient receptor potential cation channel subfamily V member 4 (TRPV4) ion channel are equally functional in the mechanotransduction of periodontal ligament cells (PDLCs) and to reveal the interplay of these mechanically sensitive ion channels (MSCs). Human PDLCs (hPDLCs) were pretreated with cytochalasin D (the inhibitor of actin polymerization), GsMTx4 (the antagonist of Piezo1) and GSK205 (the antagonist of TRPV4), and then subjected to periodic mechanical loading. The expression levels of macrophage colony stimulating factor (M-CSF), receptor activator of NF-κB ligand (RANKL) and cyclooxygenase-2 (COX2) in hPDLCs were detected via western blotting. Osteoblast mineralization induction capacity of the hPDLCs was also studied and the mitogen-activated protein kinase (MAPK) expression profile was determined via protein microarray. The expression of Piezo1 and TRPV4 in the PDLCs was significantly increased at 8 h after loading. These differences in expression were accompanied by increased expression of M-CSF, RANKL and COX2. Compared with the control group, key PDLC biomarkers were suppressed after mechanical loading following treatment with the inhibitors of Piezo1 (GsMTx4) and TRPV4 (GSK205). The phosphorylated-MAPK protein array showed differential biomarker profiles among all groups. The present study suggested that both MSCs and the cytoskeleton participated as mechanical sensors, and did so independently in hPDLC mechanotransduction. Furthermore, the Piezo1 ion channel may transmit mechanical signals via the ERK signaling pathway; however, the TRPV4 channel may function via alternative signaling pathways.
Collapse
Affiliation(s)
- Yun Shen
- Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yongchu Pan
- Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shuyu Guo
- Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lian Sun
- Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chi Zhang
- Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lin Wang
- Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
120
|
Jin P, Jan LY, Jan YN. Mechanosensitive Ion Channels: Structural Features Relevant to Mechanotransduction Mechanisms. Annu Rev Neurosci 2020; 43:207-229. [PMID: 32084327 DOI: 10.1146/annurev-neuro-070918-050509] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Activation of mechanosensitive ion channels underlies a variety of fundamental physiological processes that require sensation of mechanical force. Different mechanosensitive channels adapt distinctive structures and mechanotransduction mechanisms to fit their biological roles. How mechanosensitive channels work, especially in animals, has been extensively studied in the past decade. Here we review key findings in the functional and structural characterizations of these channels and highlight the structural features relevant to the mechanotransduction mechanism of each specific channel.
Collapse
Affiliation(s)
- Peng Jin
- Department of Physiology, University of California, San Francisco, California 94158, USA;
| | - Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, California 94158, USA; .,Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, USA
| | - Yuh-Nung Jan
- Department of Physiology, University of California, San Francisco, California 94158, USA; .,Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, USA
| |
Collapse
|
121
|
Andolfo I, Rosato BE, Manna F, De Rosa G, Marra R, Gambale A, Girelli D, Russo R, Iolascon A. Gain-of-function mutations in PIEZO1 directly impair hepatic iron metabolism via the inhibition of the BMP/SMADs pathway. Am J Hematol 2020; 95:188-197. [PMID: 31737919 DOI: 10.1002/ajh.25683] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
Dehydrated hereditary stomatocytosis (DHS), or xerocytosis, is an autosomal dominant hemolytic anemia. Most patients with DHS carry mutations in the PIEZO1 gene encoding a mechanosensitive cation channel. We here demonstrate that patients with DHS have low levels of hepcidin and only a slight increase of ERFE, the erythroid negative regulator of hepcidin. We demonstrated that at the physiological level, PIEZO1 activation induced Ca2+ influx and suppression of HAMP expression in primary hepatocytes. In two hepatic cellular models expressing PIEZO1 WT and two PIEZO1 gain-of-function mutants (R2456H and R2488Q), we highlight altered expression of a few genes/proteins involved in iron metabolism. Mutant cells showed increased intracellular Ca2+ compared to WT, which was correlated to increased phosphorylation of ERK1/2, inhibition of the BMP-SMADs pathway, and suppression of HAMP transcription. Moreover, the HuH7 cells, treated with PD0325901, a potent inhibitor of ERK1/2 phosphorylation, reduced the phosphorylation of ERK1/2 with the consequent increased phosphorylation of SMAD1/5/8, confirming the link between the two pathways. Another "proof of concept" for the mechanism that links PIEZO1 to HAMP regulation was obtained by mimicking PIEZO1 activation by cell Ca2+ overload, by the Ca2+ ionophore A23187. There was strong down-regulation of HAMP gene expression after this Ca2+ overload. Finally, the inhibition of PIEZO1 by GsMTx4 leads to phenotype rescue. This is the first demonstration of a direct link between PIEZO1 and iron metabolism, which defines the channel as a new hepatic iron metabolism regulator and as a possible therapeutic target of iron overload in DHS and other iron-loading anemias.
Collapse
Affiliation(s)
- Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Barbara Eleni Rosato
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Francesco Manna
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Gianluca De Rosa
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Roberta Marra
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Antonella Gambale
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Domenico Girelli
- Section of Internal Medicine, Department of MedicineUniversity of Verona Verona Italy
| | - Roberta Russo
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli ‘Federico II’ Naples Italy
- CEINGE, Biotecnologie Avanzate Naples Italy
| |
Collapse
|
122
|
Heng W, Bhavsar M, Han Z, Barker JH. Effects of Electrical Stimulation on Stem Cells. Curr Stem Cell Res Ther 2020; 15:441-448. [PMID: 31995020 DOI: 10.2174/1574888x15666200129154747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 11/22/2022]
Abstract
Recent interest in developing new regenerative medicine- and tissue engineering-based treatments has motivated researchers to develop strategies for manipulating stem cells to optimize outcomes in these potentially, game-changing treatments. Cells communicate with each other, and with their surrounding tissues and organs via electrochemical signals. These signals originate from ions passing back and forth through cell membranes and play a key role in regulating cell function during embryonic development, healing, and regeneration. To study the effects of electrical signals on cell function, investigators have exposed cells to exogenous electrical stimulation and have been able to increase, decrease and entirely block cell proliferation, differentiation, migration, alignment, and adherence to scaffold materials. In this review, we discuss research focused on the use of electrical stimulation to manipulate stem cell function with a focus on its incorporation in tissue engineering-based treatments.
Collapse
Affiliation(s)
- Wang Heng
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| | - Mit Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| | - Zhihua Han
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| | - John H Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Trauma & Orthopedic Surgery, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|
123
|
Mousawi F, Peng H, Li J, Ponnambalam S, Roger S, Zhao H, Yang X, Jiang LH. Chemical activation of the Piezo1 channel drives mesenchymal stem cell migration via inducing ATP release and activation of P2 receptor purinergic signaling. Stem Cells 2020; 38:410-421. [PMID: 31746084 PMCID: PMC7064961 DOI: 10.1002/stem.3114] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 08/02/2019] [Accepted: 09/01/2019] [Indexed: 12/12/2022]
Abstract
In this study, we examined the Ca2+‐permeable Piezo1 channel, a newly identified mechanosensing ion channel, in human dental pulp‐derived mesenchymal stem cells (MSCs) and hypothesized that activation of the Piezo1 channel regulates MSC migration via inducing ATP release and activation of the P2 receptor purinergic signaling. The Piezo1 mRNA and protein were readily detected in hDP‐MSCs from multiple donors and, consistently, brief exposure to Yoda1, the Piezo1 channel‐specific activator, elevated intracellular Ca2+ concentration. Yoda1‐induced Ca2+ response was inhibited by ruthenium red or GsMTx4, two Piezo1 channel inhibitors, and also by Piezo1‐specific siRNA. Brief exposure to Yoda1 also induced ATP release. Persistent exposure to Yoda1 stimulated MSC migration, which was suppressed by Piezo1‐specific siRNA, and also prevented by apyrase, an ATP scavenger, or PPADS, a P2 generic antagonist. Furthermore, stimulation of MSC migration induced by Yoda1 as well as ATP was suppressed by PF431396, a PYK2 kinase inhibitor, or U0126, an inhibitor of the mitogen‐activated protein kinase MEK/ERK signaling pathway. Collectively, these results suggest that activation of the Piezo1 channel stimulates MSC migration via inducing ATP release and subsequent activation of the P2 receptor purinergic signaling and downstream PYK2 and MEK/ERK signaling pathways, thus revealing novel insights into the molecular and signaling mechanisms regulating MSC migration. Such findings provide useful information for evolving a full understanding of MSC migration and homing and developing strategies to improve MSC‐based translational applications.
Collapse
Affiliation(s)
- Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Hongsen Peng
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Jing Li
- Lingnan Medical Research Centre, School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Sreenivasan Ponnambalam
- School of Molecular and Cell Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sébastien Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| | - Hucheng Zhao
- Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Xuebin Yang
- Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| |
Collapse
|
124
|
Wang L, You X, Lotinun S, Zhang L, Wu N, Zou W. Mechanical sensing protein PIEZO1 regulates bone homeostasis via osteoblast-osteoclast crosstalk. Nat Commun 2020; 11:282. [PMID: 31941964 PMCID: PMC6962448 DOI: 10.1038/s41467-019-14146-6] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Wolff’s law and the Utah Paradigm of skeletal physiology state that bone architecture adapts to mechanical loads. These models predict the existence of a mechanostat that links strain induced by mechanical forces to skeletal remodeling. However, how the mechanostat influences bone remodeling remains elusive. Here, we find that Piezo1 deficiency in osteoblastic cells leads to loss of bone mass and spontaneous fractures with increased bone resorption. Furthermore, Piezo1-deficient mice are resistant to further bone loss and bone resorption induced by hind limb unloading, demonstrating that PIEZO1 can affect osteoblast-osteoclast crosstalk in response to mechanical forces. At the mechanistic level, in response to mechanical loads, PIEZO1 in osteoblastic cells controls the YAP-dependent expression of type II and IX collagens. In turn, these collagen isoforms regulate osteoclast differentiation. Taken together, our data identify PIEZO1 as the major skeletal mechanosensor that tunes bone homeostasis. Mechanical forces induce bone remodeling, but how bone cells sense mechanical signaling is unclear. Here, the authors show that loss of the mechanotransduction channel Piezo1 in osteoblastic cells impairs osteoclast activity via YAP signaling and collagen expression, leading to reduced bone mass and spontaneous fractures.
Collapse
Affiliation(s)
- Lijun Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiuling You
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Sutada Lotinun
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Lingli Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Nan Wu
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China. .,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
125
|
Abstract
Stem cells can be conceptualized as computational processors capable of sensing, processing, and converting environmental information (input) to yield a specific differentiation pathway (output). In this study, we employ a temperature-controlled polymer sheet actuator to interpret and transfer information, controlled by the material’s programming, to mesenchymal stem cells. The cell’s interpretation of mechanical, thermal, and biochemical signaling is shown to be dependent on the actuator’s activity, utilized to accelerate differentiation toward bone cells, further elucidating the role of microenvironmental parameters on mammalian cells. Our method provides a unique approach to processing two discrete stimuli into one biochemical signal, calcium ions, providing a basis for the logical control of the flow of biological signals and the design of cellular functions. Stem cells are capable of sensing and processing environmental inputs, converting this information to output a specific cell lineage through signaling cascades. Despite the combinatorial nature of mechanical, thermal, and biochemical signals, these stimuli have typically been decoupled and applied independently, requiring continuous regulation by controlling units. We employ a programmable polymer actuator sheet to autonomously synchronize thermal and mechanical signals applied to mesenchymal stem cells (MSCs). Using a grid on its underside, the shape change of polymer sheet, as well as cell morphology, calcium (Ca2+) influx, and focal adhesion assembly, could be visualized and quantified. This paper gives compelling evidence that the temperature sensing and mechanosensing of MSCs are interconnected via intracellular Ca2+. Up-regulated Ca2+ levels lead to a remarkable alteration of histone H3K9 acetylation and activation of osteogenic related genes. The interplay of physical, thermal, and biochemical signaling was utilized to accelerate the cell differentiation toward osteogenic lineage. The approach of programmable bioinstructivity provides a fundamental principle for functional biomaterials exhibiting multifaceted stimuli on differentiation programs. Technological impact is expected in the tissue engineering of periosteum for treating bone defects.
Collapse
|
126
|
Xiao B. Levering Mechanically Activated Piezo Channels for Potential Pharmacological Intervention. Annu Rev Pharmacol Toxicol 2020; 60:195-218. [DOI: 10.1146/annurev-pharmtox-010919-023703] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mechanically activated Piezo channels, including Piezo1 and Piezo2 in mammals, function as key mechanotransducers for converting mechanical force into electrochemical signals. This review highlights key evidence for the potential of Piezo channel drug discovery. First, both mouse and human genetic studies have unequivocally demonstrated the prominent role of Piezo channels in various mammalian physiologies and pathophysiologies, validating their potential as novel therapeutic targets. Second, the cryo-electron microscopy structure of the 2,547-residue mouse Piezo1 trimer has been determined, providing a solid foundation for studying its structure-function relationship and drug action mechanisms and conducting virtual drug screening. Third, Piezo1 chemical activators, named Yoda1 and Jedi1/2, have been identified through high-throughput screening assays, demonstrating the drugability of Piezo channels. However, the pharmacology of Piezo channels is in its infancy. By establishing an integrated drug discovery platform, we may hopefully discover and develop a fleet of Jedi masters for battling Piezo-related human diseases.
Collapse
Affiliation(s)
- Bailong Xiao
- State Key Laboratory of Membrane Biology; Tsinghua-Peking Joint Center for Life Sciences; IDG/McGovern Institute for Brain Research; Beijing Advanced Innovation Center for Structural Biology; and School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
127
|
Sagittal Craniosynostosis with Uncommon Anatomical Pathologies in a 56-Year-Old Male Cadaver. Case Rep Pathol 2019; 2019:8034021. [PMID: 31885995 PMCID: PMC6925784 DOI: 10.1155/2019/8034021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/09/2019] [Accepted: 10/04/2019] [Indexed: 11/17/2022] Open
Abstract
Sagittal craniosynostosis (CS) is a pathologic condition that results in premature fusion of the sagittal suture, restricting the transverse growth of the skull leading in some cases to elevated intracranial pressure and neurodevelopmental delay. There is still much to be learned about the etiology of CS. Here, we report a case of 56-year-old male cadaver that we describe as sagittal CS with torus palatinus being an additional anomaly. The craniotomy was unsuccessful (cephalic index, CI = 56) and resulted in abnormal vertical outgrowth of the craniotomized bone strip. The histological analysis of the latter revealed atypical, noncompensatory massive bone overproduction. Exome sequencing of DNA extracted from the cadaveric tissue specimen performed on the Next Generation Sequencing (NGS) platform yielded 81 genetic variants identified as pathologic. Nine of those variants could be directly linked to CS with five of them targeting RhoA GTPase signaling, with a potential to make it sustained in nature. The latter could trigger upregulated calvarial osteogenesis leading to premature suture fusion, skull bone thickening, and craniotomized bone strip outgrowth observed in the present case.
Collapse
|
128
|
Hao L, Li L, Wang P, Wang Z, Shi X, Guo M, Zhang P. Synergistic osteogenesis promoted by magnetically actuated nano-mechanical stimuli. NANOSCALE 2019; 11:23423-23437. [PMID: 31799540 DOI: 10.1039/c9nr07170a] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Functional biomaterials with magnetic properties are considerably useful for regulating cell behavior and promoting bone regeneration. And the combination of such biomaterials with physical environmental cues (such as magnetic fields and mechanical stress) might be more favorable for the regulation of cell function. This study is aimed at investigating the combined effects of magnetically responsive materials and a static magnetic field (SMF) on the osteogenic differentiation of osteoblasts and the potential mechanism involved. In this study, oleic acid modified iron oxide nanoparticles (IO-OA NPs) were utilized to generate homogeneous magnetic nanocomposites with poly(lactide-co-glycolide) (PLGA) used as the base and to enhance the mechanical properties of the composites. In vitro experimental results show that in the presence of an external SMF, cell attachment and osteogenic differentiation were significantly improved using the IO-OA/PLGA composites, as indicated by enhanced alkaline phosphatase (ALP) activity, increased mineralized nodule formation, and upregulated bone-associated gene expression (ALP, OCN, and BMP2), in a dose- and time-dependent manner. Furthermore, the upregulated expression levels of piezo-type mechanosensitive ion channel component 1 (Piezo1), a key receptor for sensing mechanical stimuli, implied that the synergistically enhanced osteogenic differentiation was mainly caused as a result of the mechanical stimuli. Such magnetically actuated mechanical stimuli were induced through the nano-deformation of the magnetic substrate under a SMF, which was directly characterized via in situ scanning using atomic force microscopy (AFM). This study demonstrates that magnetically actuated nano-mechanical stimuli may underpin the synergistic effects of magnetic composites and magnetic stimuli to enhance osteogenic differentiation, and they could form the basis of a potential strategy to accelerate bone formation for bone tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Lili Hao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China. and School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| | - Linlong Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China. and University of Chinese Academy of Sciences, Beijing 100039, PR China
| | - Peng Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China. and School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China.
| | - Xincui Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China.
| | - Min Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China.
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China. and School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
129
|
Morozumi W, Inagaki S, Iwata Y, Nakamura S, Hara H, Shimazawa M. Piezo channel plays a part in retinal ganglion cell damage. Exp Eye Res 2019; 191:107900. [PMID: 31874142 DOI: 10.1016/j.exer.2019.107900] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/27/2019] [Accepted: 12/20/2019] [Indexed: 01/09/2023]
Abstract
Piezo channel is one of the mechanosensitive channels that senses pressure and shearing stress. Previous reports show that Piezo channel is expressed in many tissues such as skin and lung and they have many important roles. In addition, the mRNA of Piezo has been detected in astrocytes in the optic nerve head of mice. However, it is not yet clear where Piezo channel localize in eye and what kind of effects it have. Thus, the purpose of this study was to determine the expression sites of Piezo channel in mouse eyes and effect of Piezo channel on retinal ganglion cells. Immunostaining analysis showed that the Piezo 1/2 were expressed in the cornea, trabecular meshwork of the anterior ocular segment, lens epithelial cells, and on the retinal ganglion cell layer. The expression of retinal Piezo 2 was increased in retinal disorder model mouse caused by high IOP. Piezo 1 agonist Yoda 1 suppressed neurite outgrowth in retinal ganglion cells. On the other hand, Piezo antagonist GsMTx4 promoted neurite outgrowth in retinal ganglion cells. These findings indicate that Piezo channel may contribute to diseases relating the IOP such as glaucoma.
Collapse
Affiliation(s)
- Wataru Morozumi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoshi Inagaki
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Iwata
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
130
|
Piñeiro-Ramil M, Sanjurjo-Rodríguez C, Castro-Viñuelas R, Rodríguez-Fernández S, Fuentes-Boquete I, Blanco F, Díaz-Prado S. Usefulness of Mesenchymal Cell Lines for Bone and Cartilage Regeneration Research. Int J Mol Sci 2019; 20:E6286. [PMID: 31847077 PMCID: PMC6940884 DOI: 10.3390/ijms20246286] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/18/2022] Open
Abstract
The unavailability of sufficient numbers of human primary cells is a major roadblock for in vitro repair of bone and/or cartilage, and for performing disease modelling experiments. Immortalized mesenchymal stromal cells (iMSCs) may be employed as a research tool for avoiding these problems. The purpose of this review was to revise the available literature on the characteristics of the iMSC lines, paying special attention to the maintenance of the phenotype of the primary cells from which they were derived, and whether they are effectively useful for in vitro disease modeling and cell therapy purposes. This review was performed by searching on Web of Science, Scopus, and PubMed databases from 1 January 2015 to 30 September 2019. The keywords used were ALL = (mesenchymal AND ("cell line" OR immortal*) AND (cartilage OR chondrogenesis OR bone OR osteogenesis) AND human). Only original research studies in which a human iMSC line was employed for osteogenesis or chondrogenesis experiments were included. After describing the success of the immortalization protocol, we focused on the iMSCs maintenance of the parental phenotype and multipotency. According to the literature revised, it seems that the maintenance of these characteristics is not guaranteed by immortalization, and that careful selection and validation of clones with particular characteristics is necessary for taking advantage of the full potential of iMSC to be employed in bone and cartilage-related research.
Collapse
Affiliation(s)
- M. Piñeiro-Ramil
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
| | - C. Sanjurjo-Rodríguez
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - R. Castro-Viñuelas
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
| | - S. Rodríguez-Fernández
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
| | - I.M. Fuentes-Boquete
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| | - F.J. Blanco
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - S.M. Díaz-Prado
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain; (C.S.-R.); (R.C.-V.); (S.R.-F.)
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain;
| |
Collapse
|
131
|
Makhija EP, Espinosa-Hoyos D, Jagielska A, Van Vliet KJ. Mechanical regulation of oligodendrocyte biology. Neurosci Lett 2019; 717:134673. [PMID: 31838017 DOI: 10.1016/j.neulet.2019.134673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 12/27/2022]
Abstract
Oligodendrocytes (OL) are a subset of glial cells in the central nervous system (CNS) comprising the brain and spinal cord. The CNS environment is defined by complex biochemical and biophysical cues during development and response to injury or disease. In the last decade, significant progress has been made in understanding some of the key biophysical factors in the CNS that modulate OL biology, including their key role in myelination of neurons. Taken together, those studies offer translational implications for remyelination therapies, pharmacological research, identification of novel drug targets, and improvements in methods to generate human oligodendrocyte progenitor cells (OPCs) and OLs from donor stem cells in vitro. This review summarizes current knowledge of how various physical and mechanical cues affect OL biology and its implications for disease, therapeutic approaches, and generation of human OPCs and OLs.
Collapse
Affiliation(s)
- Ekta P Makhija
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Critical Analytics for Manufacturing Personalized-Medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, 138602, Singapore
| | - Daniela Espinosa-Hoyos
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Anna Jagielska
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| | - Krystyn J Van Vliet
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Critical Analytics for Manufacturing Personalized-Medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, 138602, Singapore; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| |
Collapse
|
132
|
Bartolozzi A, Viti F, De Stefano S, Sbrana F, Petecchia L, Gavazzo P, Vassalli M. Development of label-free biophysical markers in osteogenic maturation. J Mech Behav Biomed Mater 2019; 103:103581. [PMID: 32090910 DOI: 10.1016/j.jmbbm.2019.103581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/23/2022]
Abstract
The spatial and temporal changes of morphological and mechanical properties of living cells reflect complex functionally-associated processes. Monitoring these modifications could provide a direct information on the cellular functional state. Here we present an integrated biophysical approach to the quantification of the morphological and mechanical phenotype of single cells along a maturation pathway. Specifically, quantitative phase microscopy and single cell biomechanical testing were applied to the characterization of the maturation of human foetal osteoblasts, demonstrating the ability to identify effective label-free biomarkers along this fundamental biological process.
Collapse
Affiliation(s)
- Alice Bartolozzi
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy; Dipartimento di Ingegneria dell'Informazione, Università di Firenze, Florence, Italy
| | - Federica Viti
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy.
| | - Silvia De Stefano
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - Francesca Sbrana
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy; Schaefer South-East Europe Srl, Rovigo, Italy
| | - Loredana Petecchia
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - Paola Gavazzo
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - Massimo Vassalli
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| |
Collapse
|
133
|
Herrmann M, Jakob F. Bone Marrow Niches for Skeletal Progenitor Cells and their Inhabitants in Health and Disease. Curr Stem Cell Res Ther 2019; 14:305-319. [PMID: 30674266 DOI: 10.2174/1574888x14666190123161447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/04/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
The bone marrow hosts skeletal progenitor cells which have most widely been referred to as Mesenchymal Stem or Stromal Cells (MSCs), a heterogeneous population of adult stem cells possessing the potential for self-renewal and multilineage differentiation. A consensus agreement on minimal criteria has been suggested to define MSCs in vitro, including adhesion to plastic, expression of typical surface markers and the ability to differentiate towards the adipogenic, osteogenic and chondrogenic lineages but they are critically discussed since the differentiation capability of cells could not always be confirmed by stringent assays in vivo. However, these in vitro characteristics have led to the notion that progenitor cell populations, similar to MSCs in bone marrow, reside in various tissues. MSCs are in the focus of numerous (pre)clinical studies on tissue regeneration and repair. Recent advances in terms of genetic animal models enabled a couple of studies targeting skeletal progenitor cells in vivo. Accordingly, different skeletal progenitor cell populations could be identified by the expression of surface markers including nestin and leptin receptor. While there are still issues with the identity of, and the overlap between different cell populations, these studies suggested that specific microenvironments, referred to as niches, host and maintain skeletal progenitor cells in the bone marrow. Dynamic mutual interactions through biological and physical cues between niche constituting cells and niche inhabitants control dormancy, symmetric and asymmetric cell division and lineage commitment. Niche constituting cells, inhabitant cells and their extracellular matrix are subject to influences of aging and disease e.g. via cellular modulators. Protective niches can be hijacked and abused by metastasizing tumor cells, and may even be adapted via mutual education. Here, we summarize the current knowledge on bone marrow skeletal progenitor cell niches in physiology and pathophysiology. We discuss the plasticity and dynamics of bone marrow niches as well as future perspectives of targeting niches for therapeutic strategies.
Collapse
Affiliation(s)
- Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Wuerzburg, Wuerzburg, Germany.,Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
134
|
Blythe NM, Muraki K, Ludlow MJ, Stylianidis V, Gilbert HTJ, Evans EL, Cuthbertson K, Foster R, Swift J, Li J, Drinkhill MJ, van Nieuwenhoven FA, Porter KE, Beech DJ, Turner NA. Mechanically activated Piezo1 channels of cardiac fibroblasts stimulate p38 mitogen-activated protein kinase activity and interleukin-6 secretion. J Biol Chem 2019; 294:17395-17408. [PMID: 31586031 PMCID: PMC6873183 DOI: 10.1074/jbc.ra119.009167] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/02/2019] [Indexed: 12/03/2022] Open
Abstract
Piezo1 is a mechanosensitive cation channel with widespread physiological importance; however, its role in the heart is poorly understood. Cardiac fibroblasts help preserve myocardial integrity and play a key role in regulating its repair and remodeling following stress or injury. Here we investigated Piezo1 expression and function in cultured human and mouse cardiac fibroblasts. RT-PCR experiments confirmed that Piezo1 mRNA in cardiac fibroblasts is expressed at levels similar to those in endothelial cells. The results of a Fura-2 intracellular Ca2+ assay validated Piezo1 as a functional ion channel that is activated by its agonist, Yoda1. Yoda1-induced Ca2+ entry was inhibited by Piezo1 blockers (gadolinium and ruthenium red) and was reduced proportionally by siRNA-mediated Piezo1 knockdown or in murine Piezo1+/- cells. Results from cell-attached patch clamp recordings on human cardiac fibroblasts established that they contain mechanically activated ion channels and that their pressure responses are reduced by Piezo1 knockdown. Investigation of Yoda1 effects on selected remodeling genes indicated that Piezo1 activation increases both mRNA levels and protein secretion of IL-6, a pro-hypertrophic and profibrotic cytokine, in a Piezo1-dependent manner. Moreover, Piezo1 knockdown reduced basal IL-6 expression from cells cultured on softer collagen-coated substrates. Multiplex kinase activity profiling combined with kinase inhibitor experiments and phosphospecific immunoblotting established that Piezo1 activation stimulates IL-6 secretion via the p38 mitogen-activated protein kinase downstream of Ca2+ entry. In summary, cardiac fibroblasts express mechanically activated Piezo1 channels coupled to secretion of the paracrine signaling molecule IL-6. Piezo1 may therefore be important in regulating cardiac remodeling.
Collapse
Affiliation(s)
- Nicola M Blythe
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, 1-100 Kusumoto, Chikusa, Nagoya 464-8650, Japan
| | - Melanie J Ludlow
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Vasili Stylianidis
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht 6200MD, The Netherlands
| | - Hamish T J Gilbert
- Wellcome Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of[c27c]áBiological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester,[c27c]áM13 9PL, United Kingdom
| | - Elizabeth L Evans
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Kevin Cuthbertson
- School of Chemistry, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Richard Foster
- School of Chemistry, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of[c27c]áBiological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester,[c27c]áM13 9PL, United Kingdom
| | - Jing Li
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Mark J Drinkhill
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Frans A van Nieuwenhoven
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht 6200MD, The Netherlands
| | - Karen E Porter
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - David J Beech
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Neil A Turner
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
135
|
Sasaki F, Hayashi M, Mouri Y, Nakamura S, Adachi T, Nakashima T. Mechanotransduction via the Piezo1-Akt pathway underlies Sost suppression in osteocytes. Biochem Biophys Res Commun 2019; 521:806-813. [PMID: 31708103 DOI: 10.1016/j.bbrc.2019.10.174] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/24/2019] [Indexed: 10/25/2022]
Abstract
Osteocytes function as critical regulators of bone homeostasis by coordinating the functions of osteoblasts and osteoclasts, and are constantly exposed to mechanical force. However, the molecular mechanism underlying the mechanical signal transduction in osteocytes is not well understood. Here, we found that Yoda1, a selective Piezo1 agonist, increased intracellular calcium mobilization and dose-dependently decreased the expression of Sost (encoding Sclerostin) in the osteocytic cell line IDG-SW3. We also demonstrated that mechanical stretch of IDG-SW3 suppressed Sost expression, a result which was abrogated by treatment with the Piezo1 inhibitor GsMTx4, and the deficiency of Piezo1. Furthermore, the suppression of Sost expression was abolished by treatment with an Akt inhibitor. Taken together, these results indicate that the activation of the Piezo1-Akt pathway in osteocytes is required for mechanical stretch-induced downregulation of Sost expression.
Collapse
Affiliation(s)
- Fumiyuki Sasaki
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Mikihito Hayashi
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yuki Mouri
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Nakamura
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.
| |
Collapse
|
136
|
Wei L, Mousawi F, Li D, Roger S, Li J, Yang X, Jiang LH. Adenosine Triphosphate Release and P2 Receptor Signaling in Piezo1 Channel-Dependent Mechanoregulation. Front Pharmacol 2019; 10:1304. [PMID: 31780935 PMCID: PMC6853025 DOI: 10.3389/fphar.2019.01304] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Organs and tissues and their constituent cells are physiologically submitted to diverse types of mechanical forces or stress, one common sequence of which is release of intracellular ATP into extracellular space. Extracellular ATP is a well-established autocrine or paracrine signaling molecule that regulates multiple cell functions and mediates cell-to-cell communications via activating the purinergic P2 receptors, more specifically, ligand-gated ion channel P2X receptors and some of the G-protein-coupled P2Y receptors. The molecular mechanisms that sense mechanical and transduce forces to trigger ATP release are poorly understood. The Piezo1, a newly identified mechanosensing ion channel, shows widespread expression and confers mechanosensitivity in many different types of cells. In this mini-review, we briefly introduce the Piezo1 channel and discuss the evidence that supports its important role in the mechanoregulation of diverse cell functions and, more specifically, critical engagement of ATP release and subsequent P2 receptor activation in Piezo1 channel-dependent mechanoregulation. Such ATP release-mediated coupling of the Piezo1 channel and P2 receptors may serve a signaling mechanism that is more common than we currently understand in transducing mechanical information to regulation of the attendant cell functions in various organs and tissues.
Collapse
Affiliation(s)
- Linyu Wei
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Dongliang Li
- Department of Physiology, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Sébastien Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| | - Jing Li
- Lingnan Medical Research Centre, School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuebin Yang
- Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| |
Collapse
|
137
|
Beech DJ, Kalli AC. Force Sensing by Piezo Channels in Cardiovascular Health and Disease. Arterioscler Thromb Vasc Biol 2019; 39:2228-2239. [PMID: 31533470 PMCID: PMC6818984 DOI: 10.1161/atvbaha.119.313348] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023]
Abstract
Mechanical forces are fundamental in cardiovascular biology, and deciphering the mechanisms by which they act remains a testing frontier in cardiovascular research. Here, we raise awareness of 2 recently discovered proteins, Piezo1 and Piezo2, which assemble as transmembrane triskelions to combine exquisite force sensing with regulated calcium influx. There is emerging evidence for their importance in endothelial shear stress sensing and secretion, NO generation, vascular tone, angiogenesis, atherosclerosis, vascular permeability and remodeling, blood pressure regulation, insulin sensitivity, exercise performance, and baroreceptor reflex, and there are early suggestions of relevance to cardiac fibroblasts and myocytes. Human genetic analysis points to significance in lymphatic disease, anemia, varicose veins, and potentially heart failure, hypertension, aneurysms, and stroke. These channels appear to be versatile force sensors, used creatively to inform various force-sensing situations. We discuss emergent concepts and controversies and suggest that the potential for new important understanding is substantial.
Collapse
Affiliation(s)
- David J. Beech
- From the Department of Discovery and Translational Science, Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, England, United Kingdom
| | - Antreas C. Kalli
- From the Department of Discovery and Translational Science, Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, England, United Kingdom
| |
Collapse
|
138
|
Miyazaki A, Sugimoto A, Yoshizaki K, Kawarabayashi K, Iwata K, Kurogoushi R, Kitamura T, Otsuka K, Hasegawa T, Akazawa Y, Fukumoto S, Ishimaru N, Iwamoto T. Coordination of WNT signaling and ciliogenesis during odontogenesis by piezo type mechanosensitive ion channel component 1. Sci Rep 2019; 9:14762. [PMID: 31611621 PMCID: PMC6791893 DOI: 10.1038/s41598-019-51381-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/30/2019] [Indexed: 01/09/2023] Open
Abstract
Signal transmission from the mechanical forces to the various intracellular activities is a fundamental process during tissue development. Despite their critical role, the mechanism of mechanical forces in the biological process is poorly understood. In this study, we demonstrated that in the response to hydrostatic pressure (HP), the piezo type mechanosensitive ion channel component 1 (PIEZO1) is a primary mechanosensing receptor for odontoblast differentiation through coordination of the WNT expression and ciliogenesis. In stem cells from human exfoliated deciduous teeth (SHED), HP significantly promoted calcium deposition as well as the expression of odontogenic marker genes, PANX3 and DSPP, and WNT related-genes including WNT5b and WNT16, whereas HP inhibited cell proliferation and enhanced primary cilia expression. WNT signaling inhibitor XAV939 and primary cilia inhibitor chloral hydrate blocked the HP-induced calcium deposition. The PIEZO1 activator Yoda1 inhibited cell proliferation but induced ciliogenesis and WNT16 expression. Interestingly, HP and Yoda1 promoted nuclear translocation of RUNX2, whereas siRNA-mediated silencing of PIEZO1 decreased HP-induced nuclear translocation of RUNX2. Taken together, these results suggest that PIEZO1 functions as a mechanotransducer that connects HP signal to the intracellular signalings during odontoblast differentiation.
Collapse
Affiliation(s)
- Aya Miyazaki
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Asuna Sugimoto
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Keigo Yoshizaki
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keita Kawarabayashi
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Kokoro Iwata
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Rika Kurogoushi
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Takamasa Kitamura
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Kunihiro Otsuka
- Department of Interdisciplinary Researches for Medicine and Photonics, Institute of Post-LED Photonics, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Tomokazu Hasegawa
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Yuki Akazawa
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Tsutomu Iwamoto
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan.
| |
Collapse
|
139
|
PIEZO1 and TRPV4, which Are Distinct Mechano-Sensors in the Osteoblastic MC3T3-E1 Cells, Modify Cell-Proliferation. Int J Mol Sci 2019; 20:ijms20194960. [PMID: 31597314 PMCID: PMC6801562 DOI: 10.3390/ijms20194960] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Mechanical-loading and unloading can modify osteoblast functioning. Ca2+ signaling is one of the earliest events in osteoblasts to induce a mechanical stimulus, thereby demonstrating the importance of the underlying mechanical sensors for the sensation. Here, we examined the mechano-sensitive channels PIEZO1 and TRPV4 were involved in the process of mechano-sensation in the osteoblastic MC3T3-E1 cells. The analysis of mRNA expression revealed a high expression of Piezo1 and Trpv4 in these cells. We also found that a PIEZO1 agonist, Yoda1, induced Ca2+ response and activated cationic currents in these cells. Ca2+ response was elicited when mechanical stimulation (MS), with shear stress, was induced by fluid flow in the MC3T3-E1 cells. Gene knockdown of Piezo1 in the MC3T3-E1 cells, by transfection with siPiezo1, inhibited the Yoda1-induced response, but failed to inhibit the MS-induced response. When MC3T3-E1 cells were transfected with siTrpv4, the MS-induced response was abolished and Yoda1 response was attenuated. Moreover, the MS-induced response was inhibited by a TRPV4 antagonist HC-067047 (HC). Yoda1 response was also inhibited by HC in MC3T3-E1 cells and HEK cells, expressing both PIEZO1 and TRPV4. Meanwhile, the activation of PIEZO1 and TRPV4 reduced the proliferation of MC3T3-E1, which was reversed by knockdown of PIEZO1, and TRPV4, respectively. In conclusion, TRPV4 and PIEZO1 are distinct mechano-sensors in the MC3T3-E1 cells. However, PIEZO1 and TRPV4 modify the proliferation of these cells, implying that PIEZO1 and TRPV4 may be functional in the osteoblastic mechano-transduction. Notably, it is also found that Yoda1 can induce TRPV4-dependent Ca2+ response, when both PIEZO1 and TRPV4 are highly expressed.
Collapse
|
140
|
Abstract
How does the skeleton detect and adapt to changes in the mechanical load it has to carry?
Collapse
Affiliation(s)
- Nele Haelterman
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Joohyun Lim
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| |
Collapse
|
141
|
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
|
142
|
Ellefsen KL, Holt JR, Chang AC, Nourse JL, Arulmoli J, Mekhdjian AH, Abuwarda H, Tombola F, Flanagan LA, Dunn AR, Parker I, Pathak MM. Myosin-II mediated traction forces evoke localized Piezo1-dependent Ca 2+ flickers. Commun Biol 2019; 2:298. [PMID: 31396578 PMCID: PMC6685976 DOI: 10.1038/s42003-019-0514-3] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/18/2019] [Indexed: 02/05/2023] Open
Abstract
Piezo channels transduce mechanical stimuli into electrical and chemical signals to powerfully influence development, tissue homeostasis, and regeneration. Studies on Piezo1 have largely focused on transduction of "outside-in" mechanical forces, and its response to internal, cell-generated forces remains poorly understood. Here, using measurements of endogenous Piezo1 activity and traction forces in native cellular conditions, we show that cellular traction forces generate spatially-restricted Piezo1-mediated Ca2+ flickers in the absence of externally-applied mechanical forces. Although Piezo1 channels diffuse readily in the plasma membrane and are widely distributed across the cell, their flicker activity is enriched near force-producing adhesions. The mechanical force that activates Piezo1 arises from Myosin II phosphorylation by Myosin Light Chain Kinase. We propose that Piezo1 Ca2+ flickers allow spatial segregation of mechanotransduction events, and that mobility allows Piezo1 channels to explore a large number of mechanical microdomains and thus respond to a greater diversity of mechanical cues.
Collapse
Affiliation(s)
- Kyle L. Ellefsen
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697 USA
| | - Jesse R. Holt
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA 92697 USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697 USA
- Center for Complex Biological Systems, UC Irvine, Irvine, CA 92697 USA
| | - Alice C. Chang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305 USA
| | - Jamison L. Nourse
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA 92697 USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697 USA
| | - Janahan Arulmoli
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697 USA
- Department of Biomedical Engineering, UC Irvine, Irvine, CA 92697 USA
| | - Armen H. Mekhdjian
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305 USA
| | - Hamid Abuwarda
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA 92697 USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697 USA
| | - Francesco Tombola
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA 92697 USA
| | - Lisa A. Flanagan
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697 USA
- Department of Biomedical Engineering, UC Irvine, Irvine, CA 92697 USA
- Department of Neurology, UC Irvine, Irvine, CA 92697 USA
| | - Alexander R. Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305 USA
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305 USA
| | - Ian Parker
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697 USA
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA 92697 USA
| | - Medha M. Pathak
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA 92697 USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697 USA
- Center for Complex Biological Systems, UC Irvine, Irvine, CA 92697 USA
- Department of Biomedical Engineering, UC Irvine, Irvine, CA 92697 USA
| |
Collapse
|
143
|
Sun W, Chi S, Li Y, Ling S, Tan Y, Xu Y, Jiang F, Li J, Liu C, Zhong G, Cao D, Jin X, Zhao D, Gao X, Liu Z, Xiao B, Li Y. The mechanosensitive Piezo1 channel is required for bone formation. eLife 2019; 8:47454. [PMID: 31290742 PMCID: PMC6685704 DOI: 10.7554/elife.47454] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/06/2019] [Indexed: 12/17/2022] Open
Abstract
Mechanical load of the skeleton system is essential for the development, growth, and maintenance of bone. However, the molecular mechanism by which mechanical stimuli are converted into osteogenesis and bone formation remains unclear. Here we report that Piezo1, a bona fide mechanotransducer that is critical for various biological processes, plays a critical role in bone formation. Knockout of Piezo1 in osteoblast lineage cells disrupts the osteogenesis of osteoblasts and severely impairs bone structure and strength. Bone loss that is induced by mechanical unloading is blunted in knockout mice. Intriguingly, simulated microgravity treatment reduced the function of osteoblasts by suppressing the expression of Piezo1. Furthermore, osteoporosis patients show reduced expression of Piezo1, which is closely correlated with osteoblast dysfunction. These data collectively suggest that Piezo1 functions as a key mechanotransducer for conferring mechanosensitivity to osteoblasts and determining mechanical-load-dependent bone formation, and represents a novel therapeutic target for treating osteoporosis or mechanical unloading-induced severe bone loss.
Collapse
Affiliation(s)
- Weijia Sun
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shaopeng Chi
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Youjia Xu
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan Jiang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Caizhi Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dengchao Cao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoyan Jin
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xingcheng Gao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Bailong Xiao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
144
|
Dela Paz NG, Frangos JA. Rapid flow-induced activation of Gα q/11 is independent of Piezo1 activation. Am J Physiol Cell Physiol 2019; 316:C741-C752. [PMID: 30811222 PMCID: PMC6580164 DOI: 10.1152/ajpcell.00215.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/22/2022]
Abstract
Endothelial cell (EC) mechanochemical transduction is the process by which mechanical stimuli are sensed by ECs and transduced into biochemical signals and ultimately into physiological responses. Identifying the mechanosensor/mechanochemical transducer(s) and describing the mechanism(s) by which they receive and transmit the signals has remained a central focus within the field. The heterotrimeric G protein, Gαq/11, is proposed to be part of a macromolecular complex together with PECAM-1 at EC junctions and may constitute the mechanochemical transducer as it is rapidly activated within seconds of flow onset. The mechanically activated cation channel Piezo1 has recently been implicated due to its involvement in mediating early responses, such as calcium and ATP release. Here, we investigate the role of Piezo1 in rapid shear stress-induced Gαq/11 activation. We show that flow-induced dissociation of Gαq/11 from PECAM-1 in ECs at 15 s is abrogated by BIM-46187, a selective inhibitor of Gαq/11 activation, suggesting that Gαq/11 activation is required for PECAM-1/Gαq/11 dissociation. Although siRNA knockdown of Piezo1 caused a dramatic decrease in PECAM-1/Gαq/11 association in the basal condition, it had no effect on flow-induced dissociation. Interestingly, siRNA knockdown of Piezo1 caused a marked decrease in PECAM-1 expression. Additionally, selective blockade of Piezo1 with ion channel inhibitors had no effect on flow-induced PECAM-1/Gαq/11 dissociations. Lastly, flow onset caused increased association of Gβ1 with Piezo1 as well as with the p101 subunit of phosphoinositide 3-kinase, which were both blocked by the Gβγ inhibitor gallein. Together, our results indicate that flow-induced activation of Piezo1 is not upstream of G protein activation.
Collapse
Affiliation(s)
| | - John A Frangos
- La Jolla Bioengineering Institute , La Jolla, California
| |
Collapse
|
145
|
Sheehan K, Lee J, Chong J, Zavala K, Sharma M, Philipsen S, Maruyama T, Xu Z, Guan Z, Eilers H, Kawamata T, Schumacher M. Transcription factor Sp4 is required for hyperalgesic state persistence. PLoS One 2019; 14:e0211349. [PMID: 30811405 PMCID: PMC6392229 DOI: 10.1371/journal.pone.0211349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022] Open
Abstract
Understanding how painful hypersensitive states develop and persist beyond the initial hours to days is critically important in the effort to devise strategies to prevent and/or reverse chronic painful states. Changes in nociceptor transcription can alter the abundance of nociceptive signaling elements, resulting in longer-term change in nociceptor phenotype. As a result, sensitized nociceptive signaling can be further amplified and nocifensive behaviors sustained for weeks to months. Building on our previous finding that transcription factor Sp4 positively regulates the expression of the pain transducing channel TRPV1 in Dorsal Root Ganglion (DRG) neurons, we sought to determine if Sp4 serves a broader role in the development and persistence of hypersensitive states in mice. We observed that more than 90% of Sp4 staining DRG neurons were small to medium sized, primarily unmyelinated (NF200 neg) and the majority co-expressed nociceptor markers TRPV1 and/or isolectin B4 (IB4). Genetically modified mice (Sp4+/-) with a 50% reduction of Sp4 showed a reduction in DRG TRPV1 mRNA and neuronal responses to the TRPV1 agonist-capsaicin. Importantly, Sp4+/- mice failed to develop persistent inflammatory thermal hyperalgesia, showing a reversal to control values after 6 hours. Despite a reversal of inflammatory thermal hyperalgesia, there was no difference in CFA-induced hindpaw swelling between CFA Sp4+/- and CFA wild type mice. Similarly, Sp4+/- mice failed to develop persistent mechanical hypersensitivity to hind-paw injection of NGF. Although Sp4+/- mice developed hypersensitivity to traumatic nerve injury, Sp4+/- mice failed to develop persistent cold or mechanical hypersensitivity to the platinum-based chemotherapeutic agent oxaliplatin, a non-traumatic model of neuropathic pain. Overall, Sp4+/- mice displayed a remarkable ability to reverse the development of multiple models of persistent inflammatory and neuropathic hypersensitivity. This suggests that Sp4 functions as a critical control point for a network of genes that conspire in the persistence of painful hypersensitive states.
Collapse
Affiliation(s)
- Kayla Sheehan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Jessica Lee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Jillian Chong
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Kathryn Zavala
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Manohar Sharma
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tomoyuki Maruyama
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Zheyun Xu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Helge Eilers
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Tomoyuki Kawamata
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
146
|
Isomursu A, Lerche M, Taskinen ME, Ivaska J, Peuhu E. Integrin signaling and mechanotransduction in regulation of somatic stem cells. Exp Cell Res 2019; 378:217-225. [PMID: 30817927 DOI: 10.1016/j.yexcr.2019.01.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/10/2019] [Accepted: 01/17/2019] [Indexed: 02/06/2023]
Abstract
Somatic stem cells are characterized by their capacity for self-renewal and differentiation, making them integral for normal tissue homeostasis. Different stem cell functions are strongly affected by the specialized microenvironment surrounding the cells. Consisting of soluble signaling factors, extracellular matrix (ECM) ligands and other cells, but also biomechanical cues such as the viscoelasticity and topography of the ECM, these factors are collectively known as the niche. Cell-ECM interactions are mediated largely by integrins, a class of heterodimeric cell adhesion molecules. Integrins bind their ligands in the extracellular space and associate with the cytoskeleton inside the cell, forming a direct mechanical link between the cells and their surroundings. Indeed, recent findings have highlighted the importance of integrins in translating biophysical cues into changes in cell signaling and function, a multistep process known as mechanotransduction. The mechanical properties of the stem cell niche are important, yet the underlying molecular details of integrin-mediated mechanotransduction in stem cells, especially the roles of the different integrin heterodimers, remain elusive. Here, we introduce the reader to the concept of integrin-mediated mechanotransduction, summarize current knowledge on the role of integrin signaling and mechanotransduction in regulation of somatic stem cell functions, and discuss open questions in the field.
Collapse
Affiliation(s)
- Aleksi Isomursu
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland
| | - Martina Lerche
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland
| | - Maria E Taskinen
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland
| | - Johanna Ivaska
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland; Department of Biochemistry and Food Chemistry, University of Turku, 20520 Turku, Finland.
| | - Emilia Peuhu
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, 20520 Turku, Finland.
| |
Collapse
|
147
|
Yan L, Jiang J, Ma C, Li R, Xia Y. [Effect of knocking down Piezo1 mechanically sensitive protein on migration of MC3T3-E1 osteoblast cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:28-34. [PMID: 30644257 PMCID: PMC8337238 DOI: 10.7507/1002-1892.201806121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/10/2018] [Indexed: 01/22/2023]
Abstract
Objective To discuss the effect of Piezo1 mechanically sensitive protein in migration process of mouse MC3T3-E1 osteoblast cells. Methods The 5th-10th generation mouse MC3T3-E1 osteoblasts were divided into Piezo1-small interfering RNA (siRNA) transfection group (group A), negative control group (group B), and blank control group (group C). Piezo1-siRNA or negative control siRNA was transfected into mouse MC3T3-E1 osteoblasts by siRNA transfection reagent, respectively; group C was only added with siRNA transfection reagent; and the cell morphology was observed under inverted phase contrast microscope and fluorescence microscope, and the transfection efficiency was calculated. The expression of Piezo1 protein was detected by immunofluorescence staining and Western blot. Transwell cell migration assay and cell scratch assay were used to detect the migration of MC3T3-E1 osteoblasts after Piezo1-siRNA transfection. Results After 48 hours of transfection, group A showed a slight increase in cell volume and mutant growth, but cell colonies decreased, suspension cells increased and cell fragments increased when compared with untransfected cells. Under fluorescence microscope, green fluorescence was observed in MC3T3-E1 osteoblasts of group B, and the transfection efficiency was 68.56%±4.12%. Immunofluorescence staining and Western blot results showed that the expression level of Piezo1 protein in group A was significantly lower than that in groups B and C ( P<0.05); there was no significant difference between group B and group C ( P>0.05). Transwell cell migration assay and cell scratch assay showed that the number of cells per hole and the scratch healing rate of cells cultured for 1-4 days in group A were significantly lower than those in groups B and C ( P<0.05); there was no significant difference between group B and group C ( P>0.05). Conclusion Piezo1 knocked down by siRNA can inhibit the migration ability of MC3T3-E1 osteoblast cells.
Collapse
Affiliation(s)
- Liang Yan
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Jin Jiang
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Chongwen Ma
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Rui Li
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Yayi Xia
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000,
| |
Collapse
|
148
|
Stavenschi E, Hoey DA. Pressure-induced mesenchymal stem cell osteogenesis is dependent on intermediate filament remodeling. FASEB J 2018; 33:4178-4187. [PMID: 30550359 DOI: 10.1096/fj.201801474rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Macroscale loading of bone generates a complex local mechanical microenvironment that drives osteogenesis and bone mechanoadaptation. One such mechanical stimulus generated is hydrostatic pressure (HP); however, the effect of HP on mesenchymal stem cells (MSCs) and the mechanotransduction mechanisms utilized by these cells to sense this stimulus are yet to be fully elucidated. In this study, we demonstrate that cyclic HP is a potent mediator of cytoskeletal reorganization and increases in osteogenic responses in MSCs. In particular, we demonstrate that the intermediate filament (IF) network undergoes breakdown and reorganization with centripetal translocation of IF bundles toward the perinuclear region. Furthermore, we show for the first time that this IF remodeling is required for loading-induced MSC osteogenesis, revealing a novel mechanism of MSC mechanotransduction. In addition, we demonstrate that chemical disruption of IFs with withaferin A induces a similar mechanism of IF breakdown and remodeling as well as a subsequent increase in osteogenic gene expression in MSCs, exhibiting a potential mechanotherapeutic effect to enhance MSC osteogenesis. This study therefore highlights a novel mechanotransduction mechanism of pressure-induced MSC osteogenesis involving the understudied cytoskeletal structure, the IF, and demonstrates a potential new therapy to enhance bone formation in bone-loss diseases such as osteoporosis.-Stavenschi, E., Hoey, D. A. Pressure-induced mesenchymal stem cell osteogenesis is dependent on intermediate filament remodeling.
Collapse
Affiliation(s)
- Elena Stavenschi
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - David A Hoey
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Aeronautical and Biomedical Engineering, University of Limerick, Limerick, Ireland; and.,Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| |
Collapse
|
149
|
Mechanosensitive channels and their functions in stem cell differentiation. Exp Cell Res 2018; 374:259-265. [PMID: 30500393 DOI: 10.1016/j.yexcr.2018.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/17/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
Stem cells continuously perceive and respond to various environmental signals during development, tissue homeostasis, and pathological conditions. Mechanical force, one of the fundamental signals in the physical world, plays a vital role in the regulation of multiple functions of stem cells. The importance of cell adhesion to the extracellular matrix (ECM), cell-cell junctions, and a mechanoresponsive cell cytoskeleton has been under intensive study in the fields of stem cell biology and mechanobiology. However, the involvement of mechanosensitive (MS) ion channels in the mechanical regulation of stem cell activity has just begun to be realized. Here, we review the diversity and importance of mechanosensitive channels (MSCs), and discuss recently discovered functions of MSCs in stem cell regulation, especially in the determination of cell fate.
Collapse
|
150
|
Functional analyses of heteromeric human PIEZO1 Channels. PLoS One 2018; 13:e0207309. [PMID: 30462693 PMCID: PMC6248943 DOI: 10.1371/journal.pone.0207309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
PIEZO1 and PIEZO2 are mechanosensitive channels (MSCs) important for cellular function and mutations in them lead to human disorders. We examined how functional heteromers form between subunits of PIEZO1 using the mutants E2117K, E2117D, and E2117A. Homomers of E2117K do not conduct. E2117A homomers have low conductance with rapid inactivation, and those of E2117D have high conductance with slow inactivation. Pairing E2117K with E2117D or E2117A with E2117D gave rise to new channel species representing heteromers with distinct conductances. Whole-cell currents from co-expression of E2117A and E2117D fit well with a linear-combination model of homomeric channel currents suggesting that functional channels do not form from freely-diffusing, randomly-mixed monomers in-vitro. Whole-cell current from coexpressed PIEZO1/PIEZO2 also fit as a linear combination of homomer currents. High-resolution optical images of fluorescently-tagged channels support this interpretation because coexpressed subunits segregate into discrete domains.
Collapse
|